US20150025024A1 - Solid Compositions - Google Patents

Solid Compositions Download PDF

Info

Publication number
US20150025024A1
US20150025024A1 US14/507,267 US201414507267A US2015025024A1 US 20150025024 A1 US20150025024 A1 US 20150025024A1 US 201414507267 A US201414507267 A US 201414507267A US 2015025024 A1 US2015025024 A1 US 2015025024A1
Authority
US
United States
Prior art keywords
solid
hcv
pharmaceutically acceptable
polyoxyethylene
inhibitor
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Granted
Application number
US14/507,267
Other versions
US9034832B2 (en
Inventor
Yi Gao
Geoff Zhang
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
AbbVie Inc
Original Assignee
AbbVie Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Family has litigation
First worldwide family litigation filed litigation Critical https://patents.darts-ip.com/?family=52344053&utm_source=google_patent&utm_medium=platform_link&utm_campaign=public_patent_search&patent=US20150025024(A1) "Global patent litigation dataset” by Darts-ip is licensed under a Creative Commons Attribution 4.0 International License.
Priority claimed from US13/717,993 external-priority patent/US20130172239A1/en
Application filed by AbbVie Inc filed Critical AbbVie Inc
Priority to US14/507,267 priority Critical patent/US9034832B2/en
Publication of US20150025024A1 publication Critical patent/US20150025024A1/en
Assigned to ABBVIE INC. reassignment ABBVIE INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: GAO, YI, ZHANG, GEOFF
Application granted granted Critical
Publication of US9034832B2 publication Critical patent/US9034832B2/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/10Dispersions; Emulsions
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/40Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil
    • A61K31/4025Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil not condensed and containing further heterocyclic rings, e.g. cromakalim
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/40Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil
    • A61K31/403Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil condensed with carbocyclic rings, e.g. carbazole
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/41641,3-Diazoles
    • A61K31/41781,3-Diazoles not condensed 1,3-diazoles and containing further heterocyclic rings, e.g. pilocarpine, nitrofurantoin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/41641,3-Diazoles
    • A61K31/41841,3-Diazoles condensed with carbocyclic rings, e.g. benzimidazoles
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/439Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom the ring forming part of a bridged ring system, e.g. quinuclidine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/47Quinolines; Isoquinolines
    • A61K31/4709Non-condensed quinolines and containing further heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/4965Non-condensed pyrazines
    • A61K31/497Non-condensed pyrazines containing further heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/50Pyridazines; Hydrogenated pyridazines
    • A61K31/501Pyridazines; Hydrogenated pyridazines not condensed and containing further heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/519Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/54Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one sulfur as the ring hetero atoms, e.g. sulthiame
    • A61K31/549Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one sulfur as the ring hetero atoms, e.g. sulthiame having two or more nitrogen atoms in the same ring, e.g. hydrochlorothiazide
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/55Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having seven-membered rings, e.g. azelastine, pentylenetetrazole
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7042Compounds having saccharide radicals and heterocyclic rings
    • A61K31/7052Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides
    • A61K31/706Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides containing six-membered rings with nitrogen as a ring hetero atom
    • A61K31/7064Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides containing six-membered rings with nitrogen as a ring hetero atom containing condensed or non-condensed pyrimidines
    • A61K31/7068Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides containing six-membered rings with nitrogen as a ring hetero atom containing condensed or non-condensed pyrimidines having oxo groups directly attached to the pyrimidine ring, e.g. cytidine, cytidylic acid
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7042Compounds having saccharide radicals and heterocyclic rings
    • A61K31/7052Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides
    • A61K31/706Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides containing six-membered rings with nitrogen as a ring hetero atom
    • A61K31/7064Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides containing six-membered rings with nitrogen as a ring hetero atom containing condensed or non-condensed pyrimidines
    • A61K31/7076Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides containing six-membered rings with nitrogen as a ring hetero atom containing condensed or non-condensed pyrimidines containing purines, e.g. adenosine, adenylic acid
    • A61K31/708Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides containing six-membered rings with nitrogen as a ring hetero atom containing condensed or non-condensed pyrimidines containing purines, e.g. adenosine, adenylic acid having oxo groups directly attached to the purine ring system, e.g. guanosine, guanylic acid
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/04Peptides having up to 20 amino acids in a fully defined sequence; Derivatives thereof
    • A61K38/05Dipeptides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/04Peptides having up to 20 amino acids in a fully defined sequence; Derivatives thereof
    • A61K38/06Tripeptides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/04Peptides having up to 20 amino acids in a fully defined sequence; Derivatives thereof
    • A61K38/07Tetrapeptides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/30Macromolecular organic or inorganic compounds, e.g. inorganic polyphosphates
    • A61K47/32Macromolecular compounds obtained by reactions only involving carbon-to-carbon unsaturated bonds, e.g. carbomers, poly(meth)acrylates, or polyvinyl pyrrolidone
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/14Particulate form, e.g. powders, Processes for size reducing of pure drugs or the resulting products, Pure drug nanoparticles
    • A61K9/141Intimate drug-carrier mixtures characterised by the carrier, e.g. ordered mixtures, adsorbates, solid solutions, eutectica, co-dried, co-solubilised, co-kneaded, co-milled, co-ground products, co-precipitates, co-evaporates, co-extrudates, co-melts; Drug nanoparticles with adsorbed surface modifiers
    • A61K9/146Intimate drug-carrier mixtures characterised by the carrier, e.g. ordered mixtures, adsorbates, solid solutions, eutectica, co-dried, co-solubilised, co-kneaded, co-milled, co-ground products, co-precipitates, co-evaporates, co-extrudates, co-melts; Drug nanoparticles with adsorbed surface modifiers with organic macromolecular compounds
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/14Particulate form, e.g. powders, Processes for size reducing of pure drugs or the resulting products, Pure drug nanoparticles
    • A61K9/16Agglomerates; Granulates; Microbeadlets ; Microspheres; Pellets; Solid products obtained by spray drying, spray freeze drying, spray congealing,(multiple) emulsion solvent evaporation or extraction
    • A61K9/1605Excipients; Inactive ingredients
    • A61K9/1629Organic macromolecular compounds
    • A61K9/1635Organic macromolecular compounds obtained by reactions only involving carbon-to-carbon unsaturated bonds, e.g. polyvinyl pyrrolidone, poly(meth)acrylates

Abstract

The present invention features solid compositions comprising a selected HCV inhibitor in an amorphous form. In one embodiment, the selected HCV inhibitor is formulated in an amorphous solid dispersion which comprises a pharmaceutically acceptable hydrophilic polymer and preferably a pharmaceutically acceptable surfactant.

Description

  • This application is a continuation-in-part of U.S. patent application Ser. No. 13/717,993, filed Dec. 18, 2012, which claims priority from U.S. Provisional Patent Application Ser. No. 61/581,146, filed Dec. 29, 2011, and U.S. Provisional Patent Application Ser. No. 61/645,696, filed May 11, 2012. This application incorporates by reference the entire content of U.S. Patent Application Publication No. 2013/0172239.
  • FIELD OF THE INVENTION
  • The present invention relates to solid compositions comprising anti-HCV compounds and methods of using the same to treat HCV infection.
  • BACKGROUND
  • The hepatitis C virus (HCV) is an RNA virus belonging to the Hepacivirus genus in the Flaviviridae family. The enveloped HCV virion contains a positive stranded RNA genome encoding all known virus-specific proteins in a single, uninterrupted, open reading frame. The open reading frame comprises approximately 9500 nucleotides and encodes a single large polyprotein of about 3000 amino acids. The polyprotein comprises a core protein, envelope proteins E1 and E2, a membrane bound protein p7, and the non-structural proteins NS2, NS3, NS4A, NS4B, NS5A and NS5B.
  • HCV infection is associated with progressive liver pathology, including cirrhosis and hepatocellular carcinoma. Chronic hepatitis C may be treated with peginterferon-alpha in combination with ribavirin. Substantial limitations to efficacy and tolerability remain as many users suffer from side effects, and viral elimination from the body is often inadequate. Therefore, there is a need for new drugs to treat HCV infection.
  • SUMMARY OF THE INVENTION
  • The present invention features solid compositions comprising (1) an HCV inhibitor selected from telaprevir (VX-950), BI-201335, TMC-435 (TMC-435350), vaniprevir (MK-7009), MK-5172, asunaprevir (BMS-650032), daclatasvir (BMS-790052), danoprevir, setrobuvir (ANA-598), tegobuvir (GS-333126 or GS-9190), GS-9451, mericitabine (R-4048), IDX-184, filibuvir (PF-00868554), PSI-7977, PSI-352938, BIT-225, boceprevir, GS-5885 or GS-9256 (hereinafter a “selected HCV inhibitor”); (2) a pharmaceutically acceptable hydrophilic polymer; and optionally (3) a pharmaceutically acceptable surfactant.
  • In one aspect, the present invention features a solid composition comprising a solid dispersion, wherein the solid dispersion comprises (1) a selected HCV inhibitor in an amorphous form, (2) a pharmaceutically acceptable hydrophilic polymer, and (3) a pharmaceutically acceptable surfactant, wherein the selected HCV inhibitor is telaprevir (VX-950), BI-201335, TMC-435 (TMC-435350), vaniprevir (MK-7009), MK-5172, asunaprevir (BMS-650032), daclatasvir (BMS-790052), danoprevir, setrobuvir (ANA-598), tegobuvir (GS-333126 or GS-9190), GS-9451, mericitabine (RG-7128 or R-4048), IDX-184, filibuvir (PF-00868554), PSI-7977, PSI-352938, BIT-225, boceprevir, GS-5885 or GS-9256. The surfactant can be, without limitation, either formulated in the solid dispersion or separately combined or mixed with the solid dispersion. Preferably, the hydrophilic polymer has a Tg of at least 50° C. More preferably, the hydrophilic polymer has a Tg of at least 80° C. Highly preferably, the hydrophilic polymer has a Tg of at least 100° C. Hydrophilic polymers with Tgs of below 50° C., such as a polymer having a Tg of at least 25° C., and/or surfactants having HLB values of below 10, can also be used.
  • In one embodiment of this aspect of the invention, the hydrophilic polymer is selected from homopolymer of N-vinyl lactam, copolymer of N-vinyl lactam, cellulose ester, cellulose ether, polyalkylene oxide, polyacrylate, polymethacrylate, polyacrylamide, polyvinyl alcohol, vinyl acetate polymer, oligosaccharide, or polysaccharide. Non-limiting examples of suitable hydrophilic polymers include homopolymer of N-vinyl pyrrolidone, copolymer of N-vinyl pyrrolidone, copolymer of N-vinyl pyrrolidone and vinyl acetate, copolymer of N-vinyl pyrrolidone and vinyl propionate, graft copolymer of polyethylene glycol/polyvinyl caprolactam/polyvinyl acetate (e.g., Soluplus), polyvinylpyrrolidone, methylcellulose, ethylcellulose, hydroxyalkylcelluloses, hydroxypropylcellulose, hydroxyalkylalkylcellulose, hydroxypropylmethylcellulose, cellulose phthalate, cellulose succinate, cellulose acetate phthalate, hydroxypropylmethylcellulose phthalate, hydroxypropylmethylcellulose succinate, hydroxypropylmethylcellulose acetate succinate, polyethylene oxide, polypropylene oxide, copolymer of ethylene oxide and propylene oxide, methacrylic acid/ethyl acrylate copolymer, methacrylic acid/methyl methacrylate copolymer, butyl methacrylate/2-dimethylaminoethyl methacrylate copolymer, poly(hydroxyalkyl acrylate), poly(hydroxyalkyl methacrylate), copolymer of vinyl acetate and crotonic acid, partially hydrolyzed polyvinyl acetate, carrageenan, galactomannan, or xanthan gum, or a combination thereof. In some cases, sugar alcohols can be used in addition to, or in lieu of, hydrophilic polymers.
  • In another embodiment of this aspect of the invention, the surfactant is selected from polyoxyethylene castor oil derivates, mono fatty acid ester of polyoxyethylene sorbitan, polyoxyethylene alkyl ether, polyoxyethylene alkylaryl ether, polyethylene glycol fatty acid ester, alkylene glycol fatty acid mono ester, sucrose fatty acid ester, or sorbitan fatty acid mono ester. Non-limiting examples of suitable surfactants include polyoxyethyleneglycerol triricinoleate or polyoxyl 35 castor oil (Cremophor EL; BASF Corp.) or polyoxyethyleneglycerol oxystearate such as polyethylenglycol 40 hydrogenated castor oil (Cremophor RH 40, also known as polyoxyl 40 hydrogenated castor oil or macrogolglycerol hydroxystearate) or polyethylenglycol 60 hydrogenated castor oil (Cremophor RH 60), mono fatty acid ester of polyoxyethylene sorbitan, such as mono fatty acid ester of polyoxyethylene (20) sorbitan, e.g. polyoxyethylene (20) sorbitan monooleate (Tween 80), polyoxyethylene (20) sorbitan monostearate (Tween 60), polyoxyethylene (20) sorbitan monopalmitate (Tween 40) or polyoxyethylene (20) sorbitan monolaurate (Tween 20), polyoxyethylene (3) lauryl ether, polyoxyethylene (5) cetyl ether, polyoxyethylene (2) stearyl ether, polyoxyethylene (5) stearyl ether, polyoxyethylene (2) nonylphenyl ether, polyoxyethylene (3) nonylphenyl ether, polyoxyethylene (4) nonylphenyl ether, polyoxyethylene (3) octylphenyl ether, PEG-200 monolaurate, PEG-200 dilaurate, PEG-300 dilaurate, PEG-400 dilaurate, PEG-300 distearate, PEG-300 dioleate, propylene glycol monolaurate (e.g., lauroglycol FCC), D-alpha-tocopheryl polyethylene glycol 1000 succinate, sucrose monostearate, sucrose distearate, sucrose monolaurate, sucrose dilaurate, sorbitan mono laurate, sorbitan monooleate, sorbitan monopaInitate, or sorbitan stearate, or a combination thereof. Other suitable ionic or non-ionic surfactants may also be used.
  • In yet another embodiment of this aspect of the invention, the solid dispersion is an amorphous solid dispersion. In still another embodiment, the solid dispersion is an amorphous solid dispersion which comprises (1) the selected HCV inhibitor, (2) the hydrophilic polymer, and (3) the surfactant. In a further embodiment, the solid dispersion is a solid solution comprising (1) the selected HCV inhibitor, and (2) the hydrophilic polymer. In yet another embodiment, the solid dispersion is a solid solution comprising (1) the selected HCV inhibitor, (2) the hydrophilic polymer, and (3) the surfactant.
  • In yet another embodiment of this aspect of the invention, the hydrophilic polymer is a homopolymer or copolymer of N-vinyl pyrrolidone. Preferably, the hydrophilic polymer is copovidone.
  • In still another embodiment, the surfactant is D-alpha-tocopheryl polyethylene glycol 1000 succinate (vitamin E TPGS). In a further embodiment, the surfactant is lauroglycol FCC. In yet another embodiment, the surfactant is a combination of vitamin E TPGS and lauroglycol FCC. In still another embodiment, the surfactant is a sorbitan fatty acid ester, such as sorbitan mono laurate (Span 20). In another embodiment, the surfactant is selected from Tween 20, Tween 80, vitamin E TPGS, lauroglycol FCC, or a combination thereof.
  • In yet another embodiment, a solid composition of the invention comprises an amorphous solid dispersion or a solid solution which comprises (1) the selected HCV inhibitor, (2) copovidone, and (3) a surfactant selected from vitamin E TPGS, Span 20, or a combination thereof.
  • In another embodiment, a solid composition of the invention comprises an amorphous solid dispersion or a solid solution which comprises (1) the selected HCV inhibitor, (2) copovidone, and (3) a combination of vitamin E TPGS and lauroglycol FCC.
  • In still another embodiment, a solid composition of the invention comprises an amorphous solid dispersion or a solid solution which comprises (1) the selected HCV inhibitor, (2) copovidone, and (3) a surfactant selected from Tween 20 or Tween 80.
  • In another aspect, the present invention features processes of making a solid composition of the present invention. In one embodiment, the process comprises drying a volatile solvent in a liquid solution, wherein said solution comprises: (1) the selected HCV inhibitor; (2) a pharmaceutically acceptable hydrophilic polymer; and optionally (3) a pharmaceutically acceptable surfactant. The drying process can be carried out using any suitable solvent evaporation techniques including but not limited to spray-drying techniques.
  • In another embodiment, the process comprises solidifying a melt which comprises: (1) the selected HCV inhibitor; (2) a pharmaceutically acceptable hydrophilic polymer; and optionally (3) a pharmaceutically acceptable surfactant.
  • A solid composition of the invention may also contain other additives or ingredients, such as coloring agents, flavoring agents, lubricants or preservatives. A solid composition of the invention can be prepared into any suitable dosage forms, such as capsule, dragee, granule, powder, or tablet.
  • A solid composition of the invention may further comprise another anti-HCV agent, for example, an agent selected from HCV helicase inhibitors, HCV polymerase inhibitors, HCV protease inhibitors, HCV NS5A inhibitors, CD81 inhibitors, cyclophilin inhibitors, or internal ribosome entry site (IRES) inhibitors.
  • The present invention further features methods of using a solid composition of the present invention to treat HCV infection. The methods comprise administering a solid composition of the present invention to a patient in need thereof, thereby reducing the blood or tissue level of HCV virus in the patient.
  • Other features, objects, and advantages of the present invention are apparent in the detailed description that follows. It should be understood, however, that the detailed description, while indicating preferred embodiments of the invention, are given by way of illustration only, not limitation. Various changes and modifications within the scope of the invention will become apparent to those skilled in the art from the detailed description.
  • DETAILED DESCRIPTION
  • The present invention features solid compositions comprising (1) a selected HCV inhibitor, (2) a pharmaceutically acceptable hydrophilic polymer, and optionally (3) a pharmaceutically acceptable surfactant, wherein the selected inhibitor is telaprevir (VX-950), BI-201335, TMC-435 (TMC-435350), vaniprevir (MK-7009), MK-5172, asunaprevir (BMS-650032), daclatasvir (BMS-790052), danoprevir, setrobuvir (ANA-598), tegobuvir (GS-333126 or GS-9190), GS-9451, mericitabine (R-4048), IDX-184, filibuvir (PF-00868554), PSI-7977, PSI-352938, BIT-225, boceprevir, GS-5885 or GS-9256. Formulating the selected HCV inhibitor in an amorphous form can increase the inherent drug solubility and dissolution rate, thereby enhancing the bioavailability of the compound.
  • Telaprevir (VX-950), BI-201335, TMC-435 (TMC-435350), vaniprevir (MK-7009), MK-5172, asunaprevir (BMS-650032), danoprevir, GS-9451, boceprevir and GS-9256 are HCV protease inhibitors; daclatasvir (BMS-790052) and GS-5885 are HCV NS5A inhibitors; and setrobuvir (ANA-598), tegobuvir (GS-333126 or GS-9190), mericitabine (R-4048), IDX-184, filibuvir (PF-00868554), PSI-7977, PSI-352938 (PSI-938), and BIT-225 are polymerase inhibitors. The chemical structures of these selected HCV inhibitors are provided below:
  • Figure US20150025024A1-20150122-C00001
    Figure US20150025024A1-20150122-C00002
    Figure US20150025024A1-20150122-C00003
    Figure US20150025024A1-20150122-C00004
  • A non-limiting way to form an amorphous form of a selected HCV inhibitor described hereinabove is through the formation of solid dispersions with a polymeric carrier. The presence of hydrophilic polymer(s) and optional surfactant(s), as well as the dispersion of the selected HCV inhibitor in an amorphous form in a matrix containing the polymer(s), can significantly enhance the dissolution rate of the selected compound. In some cases, a solid dispersion formulation can also effectively maintain the selected HCV inhibitor in its supersaturation state to allow for better absorption.
  • As used herein, the term “solid dispersion” defines a system in a solid state (as opposed to a liquid or gaseous state) comprising at least two components, wherein one component is dispersed throughout the other component or components. For example, a selected HCV inhibitor described hereinabove can be dispersed in a matrix comprised of a pharmaceutically acceptable hydrophilic polymer(s) and a pharmaceutically acceptable surfactant(s). The term “solid dispersion” encompasses systems having small particles of one phase dispersed in another phase. These particles are often of less than 400 μm in size, such as less than 100, 10, or 1 μm in size. When a solid dispersion of the components is such that the system is chemically and physically uniform or homogenous throughout or consists of one phase (as defined in thermodynamics), such a solid dispersion is called a “solid solution.” A glassy solution is a solid solution in which a solute is dissolved in a glassy solvent.
  • The terms “weight percent” or “percent by weight” or “% by weight” or “wt %” denote the weight of an individual component in a composition or mixture as a percentage of the weight of the composition or mixture.
  • Modern new chemical entities tend to have higher molecular weight, greater lipophilicity and lower aqueous solubility, all of which negatively affect oral bioavailability. Despite formulation advances leading to the commercialization of enabling technologies such as lipid-based drug delivery systems (e.g. SEDDS) and nano-particles, the delivery of poorly water-soluble compounds remains challenging because of the limitations associated with each approach. Utilizing an amorphous solid dispersion (ASD) is attractive not only because it can increase the pharmacokinetic exposure of otherwise poorly absorbed drugs, but also because the final product may be delivered to the patient as a tablet or capsule, which may provide greater chemical stability and improved patient convenience compared to liquid dosage forms.
  • For all formulation approaches it is imperative to understand the intrinsic physicochemical and biopharmaceutical properties of the active drug substance prior to or at the onset of development. To that end, the biopharmaceutical classification system (BCS) has been routinely utilized to assess oral absorption and guide formulation development. For ASD formulations, the solubility/permeability of the active pharmaceutical ingredient (API) as well as the long term physical stability of the amorphous drug products are often considered. Conceptually, there are three major factors that influence the physical stability of an ASD: thermodynamic driving force (difference in drug loading and the solubility of drug in matrix), molecular mobility, and activation barrier for crystallization. The present invention relies on the use of an innovative assessment tool to rank the intrinsic physical stability of amorphous drug substances, e.g., crystallization tendency of amorphous API.
  • The molecular mobility of an amorphous material, which is often characterized by the relaxation time constant or its reciprocal, molecular mobility, is considered by many as a principal factor in determining its physical stability. Kinetic characterization of amorphous materials has been a subject of growing research in pharmaceutical field. The fact that crystallization of amorphous phases proceeds much faster in the supercooled liquid states compared to the glassy states demonstrates the importance of molecular mobility. However significant differences in crystallization tendency have been observed across compounds that cannot be explained by mobility alone. For example, some amorphous phases crystallize almost immediately at the glass temperature, Tg (e.g., progesterone, parabens, acetaminophen), some crystallize below Tg in a relatively short time (e.g., griseofulvin, nifedipine), while others are quite stable. For some of the more stable amorphous phases, crystallization in the glassy state is often not observed and it does not proceed at a significant rate above Tg without seeding. Theoretically, Tg corresponds to the temperature of which the molecular relaxation time constant of the amorphous phase is equivalent to the experimental time scale. In light of these differences it has been postulated that, in addition to mobility, the thermodynamic driving force and activation barrier to crystallization contribute to the observed physical stability differences among these compounds.
  • Shamblin et al., J. PHYS. CHEM. B 103: 4113-4121 (1999), assessed molecular mobility of amorphous materials based on heat capacity measurements and the Adam-Gibbs model. This method allows calculation of molecular mobility using temperature-modulated differential scanning calorimetry (TMDSC) that is widely available in pharmaceutical laboratories together with the Adam-Gibbs model which has been used to characterize other materials, such as polymers and ceramics.
  • Using this method, the physical stability of pharmaceutically relevant compounds can be explored in an attempt to identify thermodynamic quantities critical to crystallization. Through this analysis, the calorimetric configurational entropy has been shown to be an important factor in determining crystallization tendency above the Tg.
  • The configurational entropy typically is a measure of the difference in the number of configurations between the amorphous and the crystalline phases. For molecules in the amorphous state to crystallize, they have to pack into a specific crystal lattice with defined configuration or orientation. Therefore, higher configurational entropy values suggest a lower probability that molecules are in the desirable orientation for packing into the crystal lattice. Hence, a meta-stable amorphous compound with larger configurational entropy tends to show greater physical stability. This is consistent with the observation that large molecules with numerous rotatable bonds are often more difficult to crystallize.
  • It has been hypothesized that the configurational entropy serves as a thermodynamic measurement of the probability of nucleation while the molecular mobility dictates the rate at which a molecule can change its configurations and serves as a kinetic measurement of nucleation. Similar arguments may be applied to the rate of crystal growth as well. Therefore, these two quantities can be used to assess the intrinsic physical stability risk for the amorphous APIs.
  • Based on experimental crystallization observations of different compounds, Baird et al., J. PHARM. SCI. 99: 3787-3806 (2010); and Eerdenbrugh et al., J. PHARM. SCI. 99: 3826-3838 (2010) proposed a classification system for assessing the crystallization tendency of amorphous systems. However, crystallization experiments take relatively long time and the results are influenced by both intrinsic and extrinsic factors. The present invention utilizes the two above intrinsic properties and a different amorphous classification system (ACS) to assess the physical stability of amorphous drug candidates. The two intrinsic molecular properties can be calculated from a single convenient calorimetry measurement.
  • The structural flexibility and mobility of a molecule can be used to predict whether a compound will be kinetically stable as an amorphous phase. A physically stable amorphous API may play a role in the physical stability of a formulated ASD.
  • In the ACS used in the present invention, molecules can be categorized into four categories, as follows:
      • Class I: Stable amorphous solid/poor crystallizer, and High configurational entropy and low molecular mobility (excellent candidates for developing ASD formulations)
      • Class II: Intermediate amorphous stability/crystallizer, and High configurational entropy but high molecular mobility
      • Class III: Intermediate amorphous stability/crystallizer, and Low molecular mobility but low configurational entropy
      • Class IV: Unstable amorphous solid/good crystallizer, and Low configurational entropy and high molecular mobility (poor candidates for developing ASD formulations)
  • Mobility is highly dependent on the temperature but identical at the Tg for all glasses. Molecular mobility is usually represented by the VTF equation in the supercooled liquid state and by the AGV equation in the glassy state as follows:
  • τ ( T ) = τ 0 exp ( D T 0 T - T 0 ) ( VTF equation ) τ ( T , T f ) = τ 0 exp ( D T 0 T - ( T / T f ) T 0 ) ( AGV equation )
  • where τ is the relaxation time constant, τ0 is a constant assumed to equal to 10−14 second, D is the strength parameter, and T0 is the temperature with zero molecular mobility (τ=∞), which is called the Kauzmann temperature and is the temperature where the equilibrium supercooled liquid (i.e. ideal glass) has the same entropy as the crystalline state. Tf is the fictive temperature, which is the temperature where the ideal glass has the same configurational entropy as a real glass at a given temperature (T). It is worth noting that, by definition, at Tg the relaxation time constants are the same for all amorphous systems (i.e. τg=100 sec). The strength parameter D can be used as a convenient representation of molecular mobility at T<Tg.
  • At the glass transition temperature, Tg, the following relationship holds, which can be obtained via the VTF equation:
  • T g T 0 = D ln ( τ g / τ 0 ) + 1
  • where τg is the relaxation time constant at Tg. D and To are not independent and that Tg/T0 is a parameter associated with the strength parameter D. In many theoretical treatments, τ0 is assumed to be 10−14 sec, therefore ln(τg0)=ln(1016)=36.84 is a constant.
  • Given that Tg is the temperature associated with a constant mobility (i.e. τ=100 sec) while T0 is a temperature associated with zero mobility for ideal glasses, the ratio of Tg/T0, and therefore the value D, represent how fast the molecular mobility of an ideal glass decreases with lowering temperature. The higher the D value, the slower the rate of decrease of molecular mobility with lowering temperature, thus favors crystallization.
  • It can be further shown for ideal glasses, that:
  • ln [ τ T / τ 0 ] = D C ( T g / T ) D + C ( 1 - T g / T )
  • Where C=ln(τg0)=36.84. Given C>0, Tg/T>1, hence at a common temperature represented on the scale of Tg/T, the molecular mobility of the ideal glass is expected to be higher for a glass with larger D value. Opposite trend is true in the supercooled liquid state above Tg. Therefore the strength parameter serves a convenient indicator for molecular mobility in ideal glasses: the larger the D value, the higher the mobility (at identical Tg/T).
  • “Ideal freshly prepared glass” is one that is melt-quenched with sufficiently high cooling rates, such that no structural relaxation has occurred at temperatures below the glass transition temperature. In such “ideal freshly prepared glasses”, the fictive temperature Tf equals its glass transition temperature, Tg. Therefore molecular relaxation time constant for an “ideal freshly prepared glass” may be derived based on the AGV equation:
  • τ ( T < T g ) = τ 0 exp ( T g T · D T 0 T g - T 0 ) = τ 0 exp ( T g T · ln ( τ g / τ 0 ) )
  • The above equation demonstrates the Arrhenius behavior with regard to the temperature dependence of molecular relaxation time constants in these systems. It is further noted that, at the same value of Tg/T, the molecular relaxation time constant or mobility is the same for all “ideal freshly prepared glasses”, regardless of other characteristics of the system. At the first glance, the strength parameter does not appear to be relevant to the magnitude of molecular mobility.
  • However, configurations in real glasses are not fully arrested. Molecular motions do occur on a longer time scale which leads to structural relaxation or aging. As a result, molecular mobility of real glasses becomes a function of aging time. In reality, when a liquid is quench-cooled, structural relaxation has already occurred in any freshly prepared glass. During the process of aging, the strength parameter D plays a role in the evolution of molecular mobility, from the “ideal freshly prepared glass” where D is of no relevance, to the ideal glass where a higher D value is associated with a higher mobility. The evolution of the molecular mobility reveals a similar relationship between mobility and strength parameter, i.e. higher molecular mobility is dictated by a higher D value during this evolution process.
  • The configurational entropy at Tg would serve as a good indicator for this parameter for two reasons: (1) Amorphous pharmaceuticals are often practically stored below the glass transition temperature; (2) Configurational entropy for “ideal freshly prepared glass” is temperature independent at T<Tg.
  • During storage, the configurational entropy continuously decreases as structural relaxation occurs. However the decrease in entropy slows down with time and is far from the values in the ideal glass, even when considering the physical aging over the entire two year's of shelf-life.
  • To determine configurational entropy, instrument such as TMDSC can be calibrated to obtain accurate measurements of heat capacity. In addition, a conventional DSC scan may provide significant insight on this thermodynamic quantity. It has been observed that the change in configurational heat capacity at Tg or simply heat capacity change at Tg, ΔCp(Tg), shows a relatively good correlation with the configurational entropy and physical stability. Hence ΔCp(Tg) which can be obtained from a conventional DSC measurement, may serve as an approximate indicator or surrogate for configurational entropy. Heat capacity is a direct measurement on the modes by which a molecule can dissipate heat energies therefore is a physically meaningful measure of configurations. The heat capacity change at the glass transition temperature directly reflects the number of configurations that become available as a result of the glass-supercooled liquid transition. Because the temperature range of typical glass transition is relatively small, the contribution of anharmonic vibrations may be minimal. Therefore, such practices minimize the concerns on the true configurational origin of the excess entropy obtained via thermal analysis.
  • In addition, ΔCp(Tg) can be used to estimate the strength parameter D for a glass based on the Adam-Gibbs model and the assumption of hyperbolic temperature relationship of the configurational heat capacity, Cp conf, at temperatures above Tg:

  • K=T·C p conf ≈T g ·ΔC p(T g)
  • The entropy-based Kauzmann temperature is calculated as:
  • T 0 = T m 1 + Δ H m / K
  • where Tm and ΔHm are the temperature and enthalpy of melting, respectively. Hence the strength parameter may be derived as:
  • D = T g - T 0 T 0 · ln ( τ g / τ 0 )
  • The advantage of using ΔCp(Tg) as an estimate of configurational entropy is that this quantity can be readily measured without laborious procedures such as those required for the determination of configuration entropy. In addition, the configurational entropy at Tg may be estimated based on ΔCp(Tg) and other relevant parameters:
  • S conf ( T g ) = Δ S m - T g T m C p conf T T Δ S m - K ( 1 T g - 1 T m )
  • where ΔSm is the entropy of melting.
  • The strength parameter D can therefore be used to represent the molecular mobility of an amorphous material, and the configurational entropy can be represented by its quantity at the glass transition temperature, or more conveniently, it can be represented by the change in heat capacity at Tg, ΔCp(Tg). The high-low criterion for each quantity can then be defined to be used in the ACS assignment.
  • The criterion for stability is different across different fields of applications. Pharmaceutical products often concern the stability during the typical shelf lives, e.g., 2-3 years. A benchmarking approach may be adopted by surveying a number of pharmaceutical compounds with known physical stability, including those whose ASD formulations have been successfully commercialized. These compounds encompass a wide variety of structural features and a broad spectrum ranging from rapid crystallizers (such as acetaminophen, griseofulvin, phenobarbital, and sulfathiazole) to some that form kinetically stable amorphous phases (such as itraconazole, ketoconazole, saquinavir, ritonavir and lopinavir). These compounds include ritonavir, acetaminophen, fenofibrate, sucrose, nifedipine, griseofulvin, lopinavir, lovastatin, felodipine, indomethacin, itraconazole, ketoconazole, phenobarbital, flopropione, celecoxib, etoricoxib, rofecoxib, Valdecoxib, tolbutamide, quinidine, phenylbutazone, sulfathiazole, hydrochlorthiazide, glibenclamide, cimetidine, atropine, rac-Ibuprofen, salicin, santonin, simvastatin, and saquinavir.
  • Based on the assessments of mobility and configurational entropy, and the known physical stability for the above selected compounds in their amorphous states, the following criteria was developed:
  • (1) D≧9 as the high molecular mobility criterion;
  • (2) Sconf(Tg)R≧6 as the criterion for high configurational entropy. Alternatively high configurational entropy may be considered when ΔCp(Tg)/R≧23.
  • Choices of these criteria allow for categorization of the compounds into four categories in the context of physical stability or crystallization tendency. In many times, the configurational features of each molecule are reflected consistently by the simple measurement of ΔCp(Tg) and information can be conveniently extracted to allow the ACS determination. The use of ΔCp(Tg) allows the ACS assignment of a molecule even when no crystal form is identified, provided that the molecular mobility can be evaluated independently by other means such as viscosity measurement and the scanning rate dependence of the glass transition temperature.
  • Based on the above-described ACS model, it is believed that a selected HCV inhibitor described hereinabove is a good candidate for developing ASD formulations.
  • In one aspect, the present invention features a solid composition comprising (1) a selected HCV inhibitor, (2) a pharmaceutically acceptable hydrophilic polymer, and optionally (3) a pharmaceutically acceptable surfactant, wherein the selected HCV inhibitor is telaprevir (VX-950), BI-201335, TMC-435 (TMC-435350), vaniprevir (MK-7009), MK-5172, asunaprevir (BMS-650032), daclatasvir (BMS-790052), danoprevir, setrobuvir (ANA-598), tegobuvir (GS-333126 or GS-9190), GS-9451, mericitabine (R-4048), IDX-184, filibuvir (PF-00868554), PSI-7977, PSI-352938, BIT-225, boceprevir, GS-5885 or GS-9256. The selected HCV inhibitor and the polymer can be formulated in a solid dispersion. The surfactant may be formulated in the same solid dispersion; or the surfactant can be separately combined or mixed with the solid dispersion.
  • In one embodiment, a solid composition of the invention comprises an amorphous solid dispersion which comprises (1) the selected HCV inhibitor, (2) a pharmaceutically acceptable hydrophilic polymer, and (3) a pharmaceutically acceptable surfactant. In another embodiment, a solid composition of the invention comprises a solid solution which comprises (1) the selected HCV inhibitor, and (2) a pharmaceutically acceptable hydrophilic polymer. In still another embodiment, a solid composition of the invention comprises a solid solution which comprises (1) the selected HCV inhibitor, (2) a pharmaceutically acceptable hydrophilic polymer, and (3) a pharmaceutically acceptable surfactant. In yet another embodiment, a solid composition of the invention comprises a glassy solution which includes (1) the selected HCV inhibitor, and (2) a pharmaceutically acceptable hydrophilic polymer. In a further embodiment, a solid composition of the invention comprises a glassy solution which includes (1) the selected HCV inhibitor, (2) a pharmaceutically acceptable hydrophilic polymer, and (3) a pharmaceutically acceptable surfactant.
  • A solid composition (or a solid dispersion) of the invention can contain, for example, at least 1% by weight of the selected HCV inhibitor, preferably at least 5%, including, e.g., at least 10%. For instance, a solid composition (or a solid dispersion) of the invention can contain from 1 to 50% by weight of the selected HCV inhibitor. For another instance, a solid composition (or a solid dispersion) of the invention can contain from 5 to 30% by weight of the selected HCV inhibitor. Preferably, a solid composition (or a solid dispersion) of the invention contains from 5 to 15% by weight of the selected HCV inhibitor.
  • A solid dispersion of the invention may contain at least 30% by weight of a pharmaceutically acceptable hydrophilic polymer or a combination of such hydrophilic polymers. Preferably, the solid dispersion contains at least 40% by weight of a pharmaceutically acceptable hydrophilic polymer or a combination of such hydrophilic polymers. More preferably, the solid dispersion contains at least 50% (including, e.g., at least 60%, 70%, 80% or 90%) by weight of a pharmaceutically acceptable hydrophilic polymer or a combination of such polymers. A solid dispersion (or a solid composition) of the invention may also contain at least 1% by weight of a pharmaceutically acceptable surfactant or a combination of such surfactants. Preferably, the solid dispersion (or solid composition) contains at least 2% by weight of a pharmaceutically acceptable surfactant or a combination of such surfactants. More preferably, the solid dispersion (or solid composition) contains from 4% to 20% by weight of the surfactant(s), such as from 5% to 10% by weight of the surfactant(s).
  • In one embodiment, a solid dispersion (or a solid composition) of the invention comprises at least 30% by weight of a pharmaceutically acceptable hydrophilic polymer or a combination of such polymers, and at least 1% by weight of a pharmaceutically acceptable surfactant or a combination of such surfactants. In another embodiment, a solid dispersion (or a solid composition) of the invention comprises at least 50% by weight of a pharmaceutically acceptable hydrophilic polymer or a combination of such polymers, and from 2% to 20% by weight of a pharmaceutically acceptable surfactant or a combination of such surfactants. In yet another embodiment, a solid dispersion (or a solid composition) of the invention comprises from 50% to 90% by weight of a pharmaceutically acceptable hydrophilic polymer or a combination of such polymers, and from 3% to 15% by weight of a pharmaceutically acceptable surfactant or a combination of such surfactants. In yet another embodiment, a solid dispersion (or a solid composition) of the invention comprises from 70% to 90% by weight of a pharmaceutically acceptable hydrophilic polymer or a combination of such polymers, and from 5% to 10% by weight of a pharmaceutically acceptable surfactant or a combination of such surfactants.
  • Preferably, a hydrophilic polymer employed in the present invention has a Tg of at least 50° C., more preferably at least 60° C., and highly preferably at least 80° C. including, but not limited to from, 80° C. to 180° C., or from 100° C. to 150° C. Methods for determining Tg values of organic polymers are described in INTRODUCTION TO PHYSICAL POLYMER SCIENCE (2nd Edition by L. H. Sperling, published by John Wiley & Sons, Inc., 1992). The Tg value can be calculated as the weighted sum of the Tg values for homopolymers derived from each of the individual monomers, i.e., the polymer Tg=ΣWi·Xi where Wi is the weight percent of monomer in the organic polymer, and Xi is the Tg value for the homopolymer derived from monomer i. Tg values for the homopolymers may be taken from POLYMER HANDBOOK (2nd Edition by J. Brandrup and E. H. Immergut, Editors, published by John Wiley & Sons, Inc., 1975). Hydrophilic polymers with a Tg as described above may allow for the preparation of solid dispersions that are mechanically stable and, within ordinary temperature ranges, sufficiently temperature stable so that the solid dispersions may be used as dosage forms without further processing or be compacted to tablets with only a small amount of tabletting aids. Hydrophilic polymers having a Tg of below 50° C. may also be used.
  • Preferably, a hydrophilic polymer employed in the present invention is water-soluble. A solid composition of the present invention can also comprise poorly water-soluble or water-insoluble polymer or polymers, such as cross-linked polymers. A hydrophilic polymer comprised in a solid composition of the present invention preferably has an apparent viscosity, when dissolved at 20° C. in an aqueous solution at 2% (w/v), of 1 to 5000 mPa·s., and more preferably of 1 to 700 mPa·s, and most preferably of 5 to 100 mPa·s.
  • Hydrophilic polymers suitable for use in a solid composition of the invention include, but are not limited to, homopolymers or copolymers of N-vinyl lactams, such as homopolymers or copolymers of N-vinyl pyrrolidone (e.g., polyvinylpyrrolidone (PVP), or copolymers of N-vinyl pyrrolidone and vinyl acetate or vinyl propionate); cellulose esters or cellulose ethers, such as alkylcelluloses (e.g., methylcellulose or ethylcellulose), hydroxyalkylcelluloses (e.g., hydroxypropylcellulose), hydroxyalkylalkylcelluloses (e.g., hydroxypropylmethylcellulose), and cellulose phthalates or succinates (e.g., cellulose acetate phthalate and hydroxypropylmethylcellulose phthalate, hydroxypropylmethylcellulose succinate, or hydroxypropylmethylcellulose acetate succinate); high molecular polyalkylene oxides, such as polyethylene oxide, polypropylene oxide, and copolymers of ethylene oxide and propylene oxide; polyacrylates or polymethacrylates, such as methacrylic acid/ethyl acrylate copolymers, methacrylic acid/methyl methacrylate copolymers, butyl methacrylate/2-dimethylaminoethyl methacrylate copolymers, poly(hydroxyalkyl acrylates), and poly(hydroxyalkyl methacrylates); polyacrylamides; vinyl acetate polymers, such as copolymers of vinyl acetate and crotonic acid, and partially hydrolyzed polyvinyl acetate (also referred to as partially saponified “polyvinyl alcohol”); polyvinyl alcohol; oligo- or polysaccharides, such as carrageenans, galactomannans, and xanthan gum; polyhydroxyalkylacrylates; polyhydroxyalkyl-methacrylates; copolymers of methyl methacrylate and acrylic acid; polyethylene glycols (PEGs); graft copolymers of polyethylene glycol/polyvinyl caprolactam/polyvinyl acetate, or any mixture or combination thereof. In some cases, sugar alcohols can be used in addition to, or in lieu of, hydrophilic polymers.
  • Non-limiting examples of preferred hydrophilic polymers for the invention include polyvinylpyrrolidone (PVP) K17, PVP K25, PVP K30, PVP K90, hydroxypropyl methylcellulose (HPMC) E3, HPMC E5, HPMC E6, HPMC E15, HPMC K3, HPMC A4, HPMC A15, HPMC acetate succinate (AS) LF, HPMC AS MF, HPMC AS HF, HPMC AS LG, HPMC AS MG, HPMC AS HG, HPMC phthalate (P) 50, HPMC P 55, Ethocel 4, Ethocel 7, Ethocel 10, Ethocel 14, Ethocel 20, copovidone (vinylpyrrolidone-vinyl acetate copolymer 60/40), polyvinyl acetate, methacrylate/methacrylic acid copolymer (Eudragit) L100-55, Eudragit L100, Eudragit S100, polyethylene glycol (PEG) 400, PEG 600, PEG 1450, PEG 3350, PEG 4000, PEG 6000, PEG 8000, Soluplus, poloxamer 124, poloxamer 188, poloxamer 237, poloxamer 338, and poloxamer 407.
  • Of these, homopolymers or copolymers of N-vinyl pyrrolidone, such as copolymers of N-vinyl pyrrolidone and vinyl acetate, are preferred. A non-limiting example of a preferred polymer is a copolymer of 60% by weight of N-vinyl pyrrolidone and 40% by weight of vinyl acetate. Other preferred polymers include, without limitation, hydroxypropyl methylcellulose (HPMC, also known as hypromellose in USP), such as hydroxypropyl methylcellulose grade E5 (HPMC-E5); and hydroxypropyl methylcellulose acetate succinate (HPMC-AS).
  • A pharmaceutically acceptable surfactant employed in the present invention is preferably a non-ionic surfactant. Ionic surfactants may also be used. More preferably, a solid composition of the present invention comprises a pharmaceutically acceptable surfactant having an HLB value of from 2-20. In one example, a solid composition of the present invention includes a mixture of pharmaceutically acceptable surfactants, with at least one surfactant having an HLB value of no less than 10 and at least another surfactant having an HLB value of below 10. The HLB system (Fiedler, H. B., ENCYLOPEDIA OF EXCIPIENTS, 5th ed., Aulendorf: ECV-Editio-Cantor-Verlag (2002)) attributes numeric values to surfactants, with lipophilic substances receiving lower HLB values and hydrophilic substances receiving higher HLB values.
  • Non-limiting examples of pharmaceutically acceptable surfactants that are suitable for the present invention include polyoxyethylene castor oil derivates, e.g. polyoxyethyleneglycerol triricinoleate or polyoxyl 35 castor oil (Cremophor EL; BASF Corp.) or polyoxyethyleneglycerol oxystearate such as polyethylenglycol 40 hydrogenated castor oil (Cremophor RH 40, also known as polyoxyl 40 hydrogenated castor oil or macrogolglycerol hydroxystearate) or polyethylenglycol 60 hydrogenated castor oil (Cremophor RH 60); or a mono fatty acid ester of polyoxyethylene sorbitan, such as a mono fatty acid ester of polyoxyethylene (20) sorbitan, e.g. polyoxyethylene (20) sorbitan monooleate (Tween 80), polyoxyethylene (20) sorbitan monostearate (Tween 60), polyoxyethylene (20) sorbitan monopalmitate (Tween 40), or polyoxyethylene (20) sorbitan monolaurate (Tween 20). Other non-limiting examples of suitable surfactants include polyoxyethylene alkyl ethers, e.g. polyoxyethylene (3) lauryl ether, polyoxyethylene (5) cetyl ether, polyoxyethylene (2) stearyl ether, polyoxyethylene (5) stearyl ether; polyoxyethylene alkylaryl ethers, e.g. polyoxyethylene (2) nonylphenyl ether, polyoxyethylene (3) nonylphenyl ether, polyoxyethylene (4) nonylphenyl ether, polyoxyethylene (3) octylphenyl ether; polyethylene glycol fatty acid esters, e.g. PEG-200 monolaurate, PEG-200 dilaurate, PEG-300 dilaurate, PEG-400 dilaurate, PEG-300 distearate, PEG-300 dioleate; alkylene glycol fatty acid mono esters, e.g. propylene glycol monolaurate (lauroglycol, such as lauroglycol FCC); sucrose fatty acid esters, e.g. sucrose monostearate, sucrose distearate, sucrose monolaurate, sucrose dilaurate; sorbitan fatty acid mono esters such as sorbitan mono laurate (Span 20), sorbitan monooleate, sorbitan monopaInitate (Span 40), or sorbitan stearate; D-alpha-tocopheryl polyethylene glycol 1000 succinate; or a combination or mixture thereof. Other suitable surfactants include, but are not limited to, block copolymers of ethylene oxide and propylene oxide, also known as polyoxyethylene polyoxypropylene block copolymers or polyoxyethylene polypropyleneglycol, such as Poloxamer 124, Poloxamer 188, Poloxamer 237, Poloxamer 388, or Poloxamer 407 (BASF Wyandotte Corp.). As described above, a mixture of surfactants can be used in a solid composition of the present invention.
  • Non-limiting examples of preferred surfactants for the invention include to polysorbate 20, polysorbate 40, polysorbate 60, polysorbate 80, Cremophor RH 40, Cremophor EL, Gelucire 44/14, Gelucire 50/13, D-alpha-tocopheryl polyethylene glycol 1000 succinate (vitamin E TPGS), propylene glycol laurate, sodium lauryl sulfate, and sorbitan monolaurate.
  • In one embodiment, a solid composition of the present invention comprises an amorphous solid dispersion or solid solution which includes (1) a selected HCV inhibitor selected from telaprevir (VX-950), BI-201335, TMC-435 (TMC-435350), vaniprevir (MK-7009), MK-5172, asunaprevir (BMS-650032), daclatasvir (BMS-790052), danoprevir, setrobuvir (ANA-598), tegobuvir (GS-333126 or GS-9190), GS-9451, mericitabine (R-4048), IDX-184, filibuvir (PF-00868554), PSI-7977, PSI-352938, BIT-225, boceprevir, GS-5885 or GS-9256, and (2) a pharmaceutically acceptable hydrophilic polymer. The solid composition can also include a pharmaceutically acceptable surfactant which preferably is formulated in the amorphous solid dispersion or solid solution. The hydrophilic polymer can be selected, for example, from the group consisting of homopolymer of N-vinyl lactam, copolymer of N-vinyl lactam, cellulose ester, cellulose ether, polyalkylene oxide, polyacrylate, polymethacrylate, polyacrylamide, polyvinyl alcohol, vinyl acetate polymer, oligosaccharide, and polysaccharide. As a non-limiting example, the hydrophilic polymer is selected from the group consisting of homopolymer of N-vinyl pyrrolidone, copolymer of N-vinyl pyrrolidone, copolymer of N-vinyl pyrrolidone and vinyl acetate, copolymer of N-vinyl pyrrolidone and vinyl propionate, polyvinylpyrrolidone, methylcellulose, ethylcellulose, hydroxyalkylcelluloses, hydroxypropylcellulose, hydroxyalkylalkylcellulose, hydroxypropylmethylcellulose, cellulose phthalate, cellulose succinate, cellulose acetate phthalate, hydroxypropylmethylcellulose phthalate, hydroxypropylmethylcellulose succinate, hydroxypropylmethylcellulose acetate succinate, polyethylene oxide, polypropylene oxide, copolymer of ethylene oxide and propylene oxide, graft copolymer of polyethylene glycol/polyvinyl caprolactam/polyvinyl acetate, methacrylic acid/ethyl acrylate copolymer, methacrylic acid/methyl methacrylate copolymer, butyl methacrylate/2-dimethylaminoethyl methacrylate copolymer, poly(hydroxyalkyl acrylate), poly(hydroxyalkyl methacrylate), copolymer of vinyl acetate and crotonic acid, partially hydrolyzed polyvinyl acetate, carrageenan, galactomannan, and xanthan gum. Preferably, the hydrophilic polymer is selected from polyvinylpyrrolidone (PVP) K17, PVP K25, PVP K30, PVP K90, hydroxypropyl methylcellulose (HPMC) E3, HPMC E5, HPMC E6, HPMC E15, HPMC K3, HPMC A4, HPMC A15, HPMC acetate succinate (AS) LF, HPMC AS MF, HPMC AS HF, HPMC AS LG, HPMC AS MG, HPMC AS HG, HPMC phthalate (P) 50, HPMC P 55, Ethocel 4, Ethocel 7, Ethocel 10, Ethocel 14, Ethocel 20, copovidone (vinylpyrrolidone-vinyl acetate copolymer 60/40), polyvinyl acetate, methacrylate/methacrylic acid copolymer (Eudragit) L100-55, Eudragit L100, Eudragit S100, polyethylene glycol (PEG) 400, PEG 600, PEG 1450, PEG 3350, PEG 4000, PEG 6000, PEG 8000, Soluplus, poloxamer 124, poloxamer 188, poloxamer 237, poloxamer 338, or poloxamer 407. More preferably, the hydrophilic polymer is selected from homopolymers of vinylpyrrolidone (e.g., PVP with Fikentscher K values of from 12 to 100, or PVP with Fikentscher K values of from 17 to 30), or copolymers of 30 to 70% by weight of N-vinylpyrrolidone (VP) and 70 to 30% by weight of vinyl acetate (VA) (e.g., a copolymer of 60% by weight VP and 40% by weight VA). The surfactant can be selected, for example, from the group consisting of polyoxyethyleneglycerol triricinoleate or polyoxyl 35 castor oil (Cremophor EL; BASF Corp.) or polyoxyethyleneglycerol oxystearate, mono fatty acid ester of polyoxyethylene sorbitan, polyoxyethylene alkyl ether, polyoxyethylene alkylaryl ether, polyethylene glycol fatty acid ester, alkylene glycol fatty acid mono ester, sucrose fatty acid ester, and sorbitan fatty acid mono ester. As a non-limited example, the surfactant is selected from the group consisting of polyethylenglycol 40 hydrogenated castor oil (Cremophor RH 40, also known as polyoxyl 40 hydrogenated castor oil or macrogolglycerol hydroxystearate), polyethylenglycol 60 hydrogenated castor oil (Cremophor RH 60), a mono fatty acid ester of polyoxyethylene (20) sorbitan (e.g. polyoxyethylene (20) sorbitan monooleate (Tween 80), polyoxyethylene (20) sorbitan monostearate (Tween 60), polyoxyethylene (20) sorbitan monopalmitate (Tween 40), or polyoxyethylene (20) sorbitan monolaurate (Tween 20)), polyoxyethylene (3) lauryl ether, polyoxyethylene (5) cetyl ether, polyoxyethylene (2) stearyl ether, polyoxyethylene (5) stearyl ether, polyoxyethylene (2) nonylphenyl ether, polyoxyethylene (3) nonylphenyl ether, polyoxyethylene (4) nonylphenyl ether, polyoxyethylene (3) octylphenyl ether, PEG-200 monolaurate, PEG-200 dilaurate, PEG-300 dilaurate, PEG-400 dilaurate, PEG-300 distearate, PEG-300 dioleate, propylene glycol monolaurate, D-alpha-tocopheryl polyethylene glycol 1000 succinate, sucrose monostearate, sucrose distearate, sucrose monolaurate, sucrose dilaurate, sorbitan monolaurate, sorbitan monooleate, sorbitan monopaInitate, and sorbitan stearate. Preferably, the surfactant is selected from polysorbate 20, polysorbate 40, polysorbate 60, polysorbate 80, Cremophor RH 40, Cremophor EL, Gelucire 44/14, Gelucire 50/13, D-alpha-tocopheryl polyethylene glycol 1000 succinate (vitamin E TPGS), propylene glycol laurate, sodium lauryl sulfate, or sorbitan monolaurate. More preferably, the surfactant is selected from sorbitan monolaurate, D-alpha-tocopheryl polyethylene glycol 1000 succinate, propylene glycol monolaurate, or a combination thereof (e.g., a combination of D-alpha-tocopheryl polyethylene glycol 1000 succinate and lauroglycol FCC).
  • In another embodiment, a solid composition of the present invention comprises an amorphous solid dispersion or solid solution which includes (1) a selected HCV inhibitor described hereinabove, and (2) a homopolymer or copolymer of N-vinyl pyrrolidone (e.g., copovidone). The solid composition also comprises a pharmaceutically acceptable surfactant (e.g., vitamin E TPGS, sorbitan monolaurate, or a combination of vitamin E TPGS and lauroglycol FCC), wherein the surfactant preferably is formulated in the amorphous solid dispersion or solid solution.
  • In yet another embodiment, a solid composition of the present invention comprises an amorphous solid dispersion or solid solution which includes (1) a selected HCV inhibitor described hereinabove, (2) copovidone, and (3) a pharmaceutically acceptable surfactant (e.g., vitamin E TPGS, sorbitan monolaurate, or a combination of vitamin E TPGS and lauroglycol FCC). The amorphous solid dispersion or solid solution may also include another pharmaceutically acceptable surfactant.
  • In still another embodiment, a solid composition of the present invention comprises an amorphous solid dispersion or solid solution which includes (1) 10% by weight the selected HCV inhibitor, (2) 82% by weight copovidone, and (3) 5% by weight vitamin E TPGS and 2% by weight lauroglycol FCC. The solid composition can also include 1% by weight colloidal silica.
  • In a further embodiment, a solid composition of the present invention comprises an amorphous solid dispersion or solid solution which includes (1) 10% by weight the selected HCV inhibitor, (2) 82% by weight copovidone, and (3) 7% by weight propylene glycol monocaprylate (Capryol 90). The solid composition can also include 1% by weight colloidal silica.
  • A solid dispersion employed in the present invention preferably comprises or consists of a single-phase (defined in thermodynamics) in which the therapeutic agent(s) (e.g., a selected HCV inhibitor described hereinabove with or without another anti-HCV agent) is molecularly dispersed in a matrix containing the pharmaceutically acceptable hydrophilic polymer(s). In such cases, thermal analysis of the solid dispersion using differential scanning calorimetry (DSC) typically shows only one single Tg, and the solid dispersion does not contain any detectable crystalline HCV inhibitor as measured by X-ray powder diffraction spectroscopy.
  • A solid composition of the present invention can further include one or more other anti-HCV agents. These other anti-HCV agents can be, for example, HCV polymerase inhibitors (including nucleoside or non-nucleoside type of polymerase inhibitors), HCV protease inhibitors, HCV helicase inhibitors, CD81 inhibitors, cyclophilin inhibitors, internal ribosome entry site inhibitors, or HCV NS5A inhibitors.
  • In one embodiment, a solid composition of the invention comprises (1) a selected HCV inhibitor described hereinabove and (2) another HCV protease inhibitor. In another embodiment, a solid composition of the invention comprises (1) a selected HCV inhibitor described hereinabove, and (2) another HCV polymerase inhibitor (e.g., a non-nucleoside polymerase inhibitor, or preferably a nucleoside polymerase inhibitor). In yet another embodiment, a solid composition of the invention comprises (1) a selected HCV inhibitor described hereinabove, (2) another HCV protease inhibitor, and (3) another HCV polymerase inhibitor (e.g., a non-nucleoside polymerase inhibitor, or preferably a nucleoside polymerase inhibitor). In another embodiment, a solid composition of the invention comprises (1) a selected HCV inhibitor described hereinabove, and (2) another HCV NS5A inhibitor. In another embodiment, a solid composition of the invention comprises (1) a selected HCV inhibitor described hereinabove, (2) another HCV polymerase inhibitor (e.g., a non-nucleoside polymerase inhibitor, or preferably a nucleoside polymerase inhibitor), and (3) another HCV NS5A inhibitor. In another embodiment, a solid composition of the invention comprises (1) a selected HCV inhibitor described hereinabove, (2) another HCV protease inhibitor, and (3) another HCV NS5A inhibitor.
  • Non-limiting examples of other protease inhibitors can be selected from ACH-1095 (Achillion), ACH-1625 (Achillion), ACH-2684 (Achillion), AVL -181 (Avila), AVL-192 (Avila), BI-201335 (Boehringer Ingelheim), BMS-650032 (BMS), boceprevir, danoprevir, GS-9132 (Gilead), GS-9256 (Gilead), GS-9451 (Gilead), IDX-136 (Idenix), IDX-316 (Idenix), IDX-320 (Idenix), MK-5172 (Merck), narlaprevir, PHX-1766 (Phenomix), telaprevir, TMC-435 (Tibotec), vaniprevir, VBY708 (Virobay), VX-500 (Vertex), VX-813 (Vertex), VX-985 (Vertex), or a combination thereof. And non-limiting examples of other HCV polymerase inhibitors can be selected from ABT-072 (Abbott), ABT-333 (Abbott), ANA-598 (Anadys), BI-207127 (Boehringer Ingelheim), BILB-1941 (Boehringer Ingelheim), BMS-791325 (BMS), filibuvir, GL59728 (Glaxo), GL60667 (Glaxo), GS-9669 (Gilead), IDX-375 (Idenix), MK-3281 (Merck), tegobuvir, TMC-647055 (Tibotec), VCH-759 (Vertex & ViraChem), VCH-916 (ViraChem), VX-222 (VCH-222) (Vertex & ViraChem), VX-759 (Vertex), GS-6620 (Gilead), IDX-102 (Idenix), IDX-184 (Idenix), INX-189 (Inhibitex), MK-0608 (Merck), PSI-7977 (Pharmasset), PSI-938 (Pharmasset), RG7128 (Roche), TMC64912 (Medivir), GSK625433 (GlaxoSmithKline), BCX-4678 (BioCryst), or a combination thereof. The polymerase inhibitor may be a nucleotide polymerase inhibitor, such as GS-6620 (Gilead), IDX-102 (Idenix), IDX-184 (Idenix), INX-189 (Inhibitex), MK-0608 (Merck), PSI-7977 (Pharmasset), PSI-938 (Pharmasset), RG7128 (Roche), TMC64912 (Medivir), or a combination thereof. The polymerase inhibitor may also be a non-nucleoside polymerase inhibitor, such as ABT-072 (Abbott), ABT-333 (Abbott), ANA-598 (Anadys), BI-207127 (Boehringer Ingelheim), BILB-1941 (Boehringer Ingelheim), BMS-791325 (BMS), filibuvir, GL59728 (Glaxo), GL60667 (Glaxo), GS-9669 (Gilead), IDX-375 (Idenix), MK-3281 (Merck), tegobuvir, TMC-647055 (Tibotec), VCH-759 (Vertex & ViraChem), VCH-916 (ViraChem), VX-222 (VCH-222) (Vertex & ViraChem), VX-759 (Vertex), or a combination thereof. The present invention also contemplates the inclusion of both a nucleotide polymerase inhibitor and a non-nucleoside polymerase inhibitor in a solid composition of the invention. Non-limiting examples of other HCV NS5A inhibitors include ACH-2928 (Achillion), AZD2836 (Astra-Zeneca), AZD7295 (Astra-Zeneca), BMS-790052 (BMS), BMS-824393 (BMS), EDP-239 (Enanta), GS-5885 (Gilead), PPI-1301 (Presidio), PPI-461 (Presidio), GSK62336805, or a combination thereof.
  • A solid composition of the present invention preferably is a solid oral dosage form. Common solid oral dosage forms suitable for the present invention include, but are not limited to, capsules, dragees, granules, pills, powders and tablets, with capsules and tablets being preferred. A solid oral dosage form of the present invention can also include other excipients or inset diluents, such as sucrose, lactose or starch. Lubricants, coloring agents, releasing agents, coating agents, sweetening or flavoring agents, buffering agents, preservatives, or antioxidants can also be included in a solid oral dosage form of the present invention.
  • A solid composition of the present invention can be prepared by a variety of techniques such as, without limitation, melt-extrusion, spray-drying, co-precipitation, freeze drying, or other solvent evaporation techniques, with melt-extrusion and spray-drying being preferred. The melt-extrusion process typically comprises the steps of preparing a melt which includes the active ingredient(s), the hydrophilic polymer(s) and preferably the surfactant(s), and then cooling the melt until it solidifies. Melting often involves a transition into a liquid state in which it is possible for one component to get dissolved or embedded, preferably homogeneously dissolved or embedded, in the other component or components. In many cases, the polymer component(s) will melt and the other components including the active ingredient(s) and surfactant(s) will dissolve in the melt thereby forming a solution. In such a case, the polymer functions as a solvent. Melting usually involves heating above the softening point of the polymer(s). The preparation of the melt can take place in a variety of ways. The mixing of the components can take place before, during or after the formation of the melt. For example, the components can be mixed first and then melted or be simultaneously mixed and melted. The melt can also be homogenized in order to disperse the active ingredient(s) efficiently. In addition, it may be convenient first to melt the polymer(s) and then to mix in and homogenize the active ingredient(s). In one example, all materials except surfactant(s) are blended and fed into an extruder, while the surfactant(s) is molten externally and pumped in during extrusion.
  • In another example, the melt comprises a selected HCV inhibitor described hereinabove, and one or more hydrophilic polymers described above; and the melt temperature is in the range of from 100 to 170° C., preferably from 120 to 150° C., and highly preferably from 135 to 140° C. The melt can also include a pharmaceutically acceptable surfactant described above.
  • In still another example, the melt comprises a selected HCV inhibitor described hereinabove, at least another anti-HCV agent described above, and one or more hydrophilic polymers described above. The melt can also include a pharmaceutically acceptable surfactant described above.
  • To start a melt-extrusion process, the active ingredient(s) (e.g., a selected HCV inhibitor described hereinabove) can be employed in their solid forms, such as their respective crystalline forms. The active ingredient(s) can also be employed as a solution or dispersion in a suitable liquid solvent such as alcohols, aliphatic hydrocarbons, esters or, in some cases, liquid carbon dioxide. The solvent can be removed, e.g. evaporated, upon preparation of the melt.
  • Various additives can also be included in the melt, for example, flow regulators (e.g., colloidal silica), binders, lubricants, fillers, disintegrants, plasticizers, colorants, or stabilizers (e.g., antioxidants, light stabilizers, radical scavengers, and stabilizers against microbial attack).
  • The melting and/or mixing can take place in an apparatus customary for this purpose. Particularly suitable ones are extruders or kneaders. Suitable extruders include single screw extruders, intermeshing screw extruders or multiscrew extruders, preferably twin screw extruders, which can be corotating or counterrotating and, optionally, be equipped with kneading disks. It will be appreciated that the working temperatures will be determined by the kind of extruder or the kind of configuration within the extruder that is used. Part of the energy needed to melt, mix and dissolve the components in the extruder can be provided by heating elements. However, the friction and shearing of the material in the extruder may also provide a substantial amount of energy to the mixture and aid in the formation of a homogeneous melt of the components.
  • The melt can range from thin to pasty to viscous. Shaping of the extrudate can be conveniently carried out by a calender with two counter-rotating rollers with mutually matching depressions on their surface. The extrudate can be cooled and allow to solidify. The extrudate can also be cut into pieces, either before (hot-cut) or after solidification (cold-cut).
  • The solidified extrusion product can be further milled, ground or otherwise reduced to granules. The solidified extrudate, as well as each granule produced, comprises a solid dispersion, preferably a solid solution, of the active ingredient(s) in a matrix comprised of the hydrophilic polymer(s) and optionally the pharmaceutically acceptable surfactant(s). Where the granules do not contain any surfactant, a pharmaceutically acceptable surfactant described above can be added to and blended with the granules. The extrusion product can also be blended with other active ingredient(s) and/or additive(s) before being milled or ground to granules. The granules can be further processed into suitable solid oral dosage forms.
  • In some cases, direct-shaping techniques such as injection moulding can be used in combination with melt extrusion to prepare suitable solid dosage forms.
  • In one example, copovidone and one or more surfactants are mixed and granulated, followed by the addition of aerosil and a selected HCV inhibitor described hereinabove. The mixture, which may contain for example at least 5% by weight of the selected HCV inhibitor is then milled. The mixture is then subject to extrusion, and the extrudate thus produced can be milled and sieved for further processing to make capsules or tablets. Surfactant(s) employed in this example can also be added through liquid dosing during extrusion.
  • The approach of solvent evaporation, via spray-drying, provides the advantage of allowing for processability at lower temperatures, if needed, and allows for other modifications to the process in order to further improve powder properties. The spray-dried powder can then be formulated further, if needed, and final drug product is flexible with regards to whether capsule, tablet or any other solid dosage form is desired.
  • Exemplary spray-drying processes and spray-drying equipment are described in K. Masters, SPRAY DRYING HANDBOOK (Halstead Press, New York, 4th ed., 1985). Non-limiting examples of spray-drying devices that are suitable for the present invention include spray dryers manufactured by Niro Inc. or GEA Process Engineering Inc., Buchi Labortechnik AG, and Spray Drying Systems, Inc. A spray-drying process generally involves breaking up a liquid mixture into small droplets and rapidly removing solvent from the droplets in a container (spray drying apparatus) where there is a strong driving force for evaporation of solvent from the droplets. Atomization techniques include, for example, two-fluid or pressure nozzles, or rotary atomizers. The strong driving force for solvent evaporation can be provided, for example, by maintaining the partial pressure of solvent in the spray drying apparatus well below the vapor pressure of the solvent at the temperatures of the drying droplets. This may be accomplished by either (1) maintaining the pressure in the spray drying apparatus at a partial vacuum; (2) mixing the liquid droplets with a warm drying gas (e.g., heated nitrogen); or (3) both.
  • The temperature and flow rate of the drying gas, as well as the spray dryer design, can be selected so that the droplets are dry enough by the time they reach the wall of the apparatus. This help to ensure that the dried droplets are essentially solid and can form a fine powder and do not stick to the apparatus wall. The spray-dried product can be collected by removing the material manually, pneumatically, mechanically or by other suitable means. The actual length of time to achieve the preferred level of dryness depends on the size of the droplets, the formulation, and spray dryer operation. Following the solidification, the solid powder may stay in the spray drying chamber for additional time (e.g., 5-60 seconds) to further evaporate solvent from the solid powder. The final solvent content in the solid dispersion as it exits the dryer is preferably at a sufficiently low level so as to improve the stability of the final product. For instance, the residual solvent content of the spray-dried powder can be less than 2% by weight. Highly preferably, the residual solvent content is within the limits set forth in the International Conference on Harmonization (ICH) Guidelines. In addition, it may be useful to subject the spray-dried composition to further drying to lower the residual solvent to even lower levels. Methods to further lower solvent levels include, but are not limited to, fluid bed drying, infra-red drying, tumble drying, vacuum drying, and combinations of these and other processes.
  • Like the solid extrudate described above, the spray dried product contains a solid dispersion, preferably a solid solution, of the active ingredient(s) in a matrix comprised of the hydrophilic polymer(s) and optionally the pharmaceutically acceptable surfactant(s). Where the spray dried product does not contain any surfactant, a pharmaceutically acceptable surfactant described above can be added to and blended with the spray-dried product before further processing.
  • Before feeding into a spray dryer, the active ingredient(s) (e.g., a selected HCV inhibitor described hereinabove), the hydrophilic polymer(s), as well as other optional active ingredients or excipients such as the pharmaceutically acceptable surfactant(s), can be dissolved in a solvent. Suitable solvents include, but are not limited to, water, alkanols (e.g., methanol, ethanol, 1-propanol, 2-propanol or mixtures thereof), acetone, acetone/water, alkanol/water mixtures (e.g., ethanol/water mixtures), or combinations thereof. The solution can also be preheated before being fed into the spray dryer.
  • The solid dispersion produced by melt-extrusion, spray-drying or other techniques can be prepared into any suitable solid oral dosage forms. In one embodiment, the solid dispersion prepared by melt-extrusion, spray-drying or other techniques (e.g., the extrudate or the spray-dried powder) can be compressed into tablets. The solid dispersion can be either directly compressed, or milled or ground to granules or powders before compression. Compression can be done in a tablet press, such as in a steel die between two moving punches. When a solid composition of the present invention comprises a selected HCV inhibitor described hereinabove and another anti-HCV agent, it is possible to separately prepare solid dispersions of each individual active ingredient and then blend the optionally milled or ground solid dispersions before compacting. A selected HCV inhibitor described hereinabove and other active ingredient(s) can also be prepared in the same solid dispersion, optionally milled and/or blended with other additives, and then compressed into tablets.
  • At least one additive selected from flow regulators, binders, lubricants, fillers, disintegrants, or plasticizers may be used in compressing the solid dispersion. These additives can be mixed with ground or milled solid dispersion before compacting. Disintegrants promote a rapid disintegration of the compact in the stomach and keeps the liberated granules separate from one another. Non-limiting examples of suitable disintegrants are cross-linked polymers such as cross-linked polyvinyl pyrrolidone, cross-linked sodium carboxymethylcellulose or sodium croscarmellose. Non-limiting examples of suitable fillers (also referred to as bulking agents) are lactose monohydrate, calcium hydrogenphosphate, microcrystalline cellulose (e.g., Avicell), silicates, in particular silicium dioxide, magnesium oxide, talc, potato or corn starch, isomalt, or polyvinyl alcohol. Non-limiting examples of suitable flow regulators include highly dispersed silica (e.g., colloidal silica such as Aerosil), and animal or vegetable fats or waxes. Non-limiting examples of suitable lubricants include polyethylene glycol (e.g., having a molecular weight of from 1000 to 6000), magnesium and calcium stearates, sodium stearyl fumarate, and the like.
  • Various other additives may also be used in preparing a solid composition of the present invention, for example dyes such as azo dyes, organic or inorganic pigments such as aluminium oxide or titanium dioxide, or dyes of natural origin; stabilizers such as antioxidants, light stabilizers, radical scavengers, stabilizers against microbial attack.
  • Solid compositions according to certain embodiments of the present invention may contain several layers, for example laminated or multilayer tablets. They can be in open or closed form. “Closed dosage forms” are those in which one layer is completely surrounded by at least one other layer.
  • In order to facilitate the intake of a solid dosage form, it is advantageous to give the dosage form an appropriate shape. Large tablets that can be swallowed comfortably are therefore preferably elongated rather than round in shape.
  • A film coat on the tablet further contributes to the ease with which it can be swallowed. A film coat also improves taste and provides an elegant appearance. The film-coat usually includes a polymeric film-forming material such as hydroxypropyl methylcellulose, hydroxypropylcellulose, and acrylate or methacrylate copolymers. Besides a film-forming polymer, the film-coat may further comprise a plasticizer, e.g. polyethylene glycol, a surfactant, e.g. polysorbates, and optionally a pigment, e.g. titanium dioxide or iron oxides. The film-coating may also comprise talc as anti-adhesive. Preferably, the film coat accounts for less than 5% by weight of a pharmaceutical composition of the present invention.
  • In another aspect, the present invention feature methods of using solid compositions of the present invention to treat HIV infection. The methods comprise administering a solid composition of the present invention to a patient in need thereof. A solid composition of the present invention can be administered either alone, or in combination with one or more other anti-HCV agents, such as those described hereinabove. The specific inhibitory dose for any particular patient will depend upon a variety of factors including the severity of the HCV infection; the activity of the active ingredient(s) in the particular patient; the specific solid composition employed; the age, body weight, general health, sex and diet of the patient; the time of administration and rate of excretion; the duration of the treatment; drugs used in combination or coincidental with the selected HCV inhibitor described hereinabove; and like factors well known in the medical arts.
  • In one embodiment, a method of the present invention comprises administering to a patient in need thereof a solid composition of the present invention and at least another anti-HCV agent, wherein said another anti-HCV agent is selected from HCV polymerase inhibitors (e.g., nucleoside or non-nucleoside HCV polymerase inhibitors), HCV protease inhibitors, HCV helicase inhibitors, CD81 inhibitors, cyclophilin inhibitors, internal ribosome entry site inhibitors, or HCV NS5A inhibitors. Preferably, said another anti-HCV agent is an HCV polymerase inhibitor (e.g., nucleoside or non-nucleoside HCV polymerase inhibitor) or an HCV protease inhibitor. Also preferably, said another anti-HCV agent is interferon or ribavirin, or preferably a combination thereof. The interferon preferably is α-interferon, and more preferably, pegylated interferon-α such as PEGASYS (peginterferon alfa-2a). The administration of a solid composition of the present invention and another anti-HCV agent(s) can be concurrent or sequential.
  • The present invention also features use of a solid composition of the present invention for the manufacture of medicaments for the treatment of HCV infection.
  • In one embodiment, the selected HCV inhibitor used in any aspect, embodiment, example or feature described hereinabove is telaprevir (VX-950).
  • In another embodiment, the selected HCV inhibitor used in any aspect, embodiment, example or feature described hereinabove is BI-201335.
  • In another embodiment, the selected HCV inhibitor used in any aspect, embodiment, example or feature described hereinabove is TMC-435 (TMC-435350).
  • In another embodiment, the selected HCV inhibitor used in any aspect, embodiment, example or feature described hereinabove is vaniprevir (MK-7009).
  • In another embodiment, the selected HCV inhibitor used in any aspect, embodiment, example or feature described hereinabove is MK-5172.
  • In another embodiment, the selected HCV inhibitor used in any aspect, embodiment, example or feature described hereinabove is asunaprevir (BMS-650032).
  • In another embodiment, the selected HCV inhibitor used in any aspect, embodiment, example or feature described hereinabove is daclatasvir (BMS-790052).
  • In another embodiment, the selected HCV inhibitor used in any aspect, embodiment, example or feature described hereinabove is danoprevir. Preferably, danoprevir is used together with ritonavir to improve the pharmacokinetics of danoprevir. More preferably, danoprevir is co-formulated with ritonavir in a solid composition of the invention. For instance, danoprevir and ritonavir in a solid composition of the invention can be formulated in the same solid dispersion or different solid dispersions.
  • In another embodiment, the selected HCV inhibitor used in any aspect, embodiment, example or feature described hereinabove is setrobuvir (ANA-598).
  • In another embodiment, the selected HCV inhibitor used in any aspect, embodiment, example or feature described hereinabove is tegobuvir (GS-333126 or GS-9190).
  • Preferably, a solid composition of this embodiment further comprises GS-9256, GS-9451 or GS-5885. Also preferably, a solid composition of this embodiment further comprises GS-9451 and GS-5885.
  • In another embodiment, the selected HCV inhibitor used in any aspect, embodiment, example or feature described hereinabove is GS-9451. Preferably, a solid composition of this embodiment further comprises tegobuvir or GS-5885. Also preferably, a solid composition of this embodiment further comprises tegobuvir and GS-5885.
  • In another embodiment, the selected HCV inhibitor used in any aspect, embodiment, example or feature described hereinabove is mericitabine (R-4048).
  • In another embodiment, the selected HCV inhibitor used in any aspect, embodiment, example or feature described hereinabove is IDX-184.
  • In another embodiment, the selected HCV inhibitor used in any aspect, embodiment, example or feature described hereinabove is filibuvir (PF-00868554).
  • In another embodiment, the selected HCV inhibitor used in any aspect, embodiment, example or feature described hereinabove is PSI-7977. Preferably, a solid composition of this embodiment further comprises GS-5885 or daclatasvir.
  • In another embodiment, the selected HCV inhibitor used in any aspect, embodiment, example or feature described hereinabove is PSI-352938.
  • In another embodiment, the selected HCV inhibitor used in any aspect, embodiment, example or feature described hereinabove is BIT-225.
  • In another embodiment, the selected HCV inhibitor used in any aspect, embodiment, example or feature described hereinabove is boceprevir.
  • In another embodiment, the selected HCV inhibitor used in any aspect, embodiment, example or feature described hereinabove is GS-5885. Preferably, a solid composition of this embodiment further comprises PSI-7977, GS-9451 or tegobuvir. Also preferably, a solid composition of this embodiment further comprises GS-9451 and tegobuvir. In one example, a solid oral dosage form of the invention comprises an amorphous solid dispersion, wherein said solid dispersion comprises GS-5885, copovidone and optionally a pharmaceutically acceptable surfactant. In another example, a solid oral dosage form of the invention comprises an amorphous solid dispersion, wherein said solid dispersion comprises GS-5885, copovidone and optionally a pharmaceutically acceptable surfactant, and wherein said solid oral dosage form further comprises
  • Figure US20150025024A1-20150122-C00005
  • In another embodiment, the selected HCV inhibitor used in any aspect, embodiment, example or feature described hereinabove is GS-9256. Preferably, a solid composition of this embodiment further comprises tegobuvir.
  • Other formulation approaches, such as liquid-based formulations, simple solutions, nanoparticles, crystalline solids, salts or co-crystals, and conventional immediate release formulations, can also be employed to formulate the selected HCV inhibitors, either alone or in combination with other anti-HCV agents.
  • The foregoing description of the present invention provides illustration and description, but is not intended to be exhaustive or to limit the invention to the precise one disclosed. Modifications and variations are possible in light of the above teachings or may be acquired from practice of the invention. Thus, it is noted that the scope of the invention is defined by the claims and their equivalents.

Claims (2)

What is claimed is:
1. A solid oral dosage form comprising an amorphous solid dispersion which includes:
Figure US20150025024A1-20150122-C00006
copovidone, and
optionally a pharmaceutically acceptable surfactant.
2. The solid oral dosage form of claim 1, further comprising
Figure US20150025024A1-20150122-C00007
US14/507,267 2011-12-29 2014-10-06 Solid compositions Active US9034832B2 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US14/507,267 US9034832B2 (en) 2011-12-29 2014-10-06 Solid compositions

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US201161581146P 2011-12-29 2011-12-29
US201261645696P 2012-05-11 2012-05-11
US13/717,993 US20130172239A1 (en) 2011-12-29 2012-12-18 Solid compositions
US14/507,267 US9034832B2 (en) 2011-12-29 2014-10-06 Solid compositions

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US13/717,993 Continuation-In-Part US20130172239A1 (en) 2011-12-29 2012-12-18 Solid compositions

Publications (2)

Publication Number Publication Date
US20150025024A1 true US20150025024A1 (en) 2015-01-22
US9034832B2 US9034832B2 (en) 2015-05-19

Family

ID=52344053

Family Applications (1)

Application Number Title Priority Date Filing Date
US14/507,267 Active US9034832B2 (en) 2011-12-29 2014-10-06 Solid compositions

Country Status (1)

Country Link
US (1) US9034832B2 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN113209087A (en) * 2020-02-05 2021-08-06 歌礼药业(浙江)有限公司 Pharmaceutical composition for inhibiting coronavirus and application thereof

Families Citing this family (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
NZ625087A (en) 2013-01-31 2017-05-26 Gilead Pharmasset Llc Combination formulation of two antiviral compounds
ES2900570T3 (en) 2013-08-27 2022-03-17 Gilead Pharmasset Llc Combination formulation of two antiviral compounds
TWI721947B (en) 2014-06-11 2021-03-21 美商基利法瑪席特有限責任公司 Solid forms of an antiviral compound

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2010017432A1 (en) * 2008-08-07 2010-02-11 Schering Corporation Pharmaceutical formulations of an hcv protease inhibitor in a solid molecular dispersion
WO2010097229A2 (en) * 2009-02-27 2010-09-02 Ortho-Mcneil-Janssen Pharmaceuticals Inc Amorphous salt of a macrocyclic inhibitor of hcv
US20120172290A1 (en) * 2009-06-11 2012-07-05 Abbott Laboratories Anti-Viral Compounds

Family Cites Families (203)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
DE75755C (en) 1894-06-14 DAHL & COMP, in Barmen Process for the preparation of aromatically substituted amidodinaphthyl methanes.
AU7044994A (en) 1993-05-24 1994-12-20 Nycomed Pharma Hemoregulatory peptides
SI9500173B (en) 1995-05-19 2002-02-28 Lek, Three-phase pharmaceutical form with constant and controlled release of amorphous active ingredient for single daily application
US6037157A (en) 1995-06-29 2000-03-14 Abbott Laboratories Method for improving pharmacokinetics
US5935982A (en) 1997-02-28 1999-08-10 The University Of North Carolina At Chapel Hill Methods of treating retroviral infection and compounds useful therefor
US6235493B1 (en) 1997-08-06 2001-05-22 The Regents Of The University Of California Amino acid substituted-cresyl violet, synthetic fluorogenic substrates for the analysis of agents in individual in vivo cells or tissue
IL139826A0 (en) 1998-05-22 2002-02-10 Avanir Pharmaceuticals BENZIMIDAZOLE DERIVATIVES AS MODULATORS OF IgE
US6369091B1 (en) 1998-05-22 2002-04-09 Avanir Pharmaceuticals Benzimidazole analogs as down-regulators of IgE
US6911462B2 (en) 1998-05-22 2005-06-28 Avanir Pharmaceuticals Benzimidazole compounds for regulating IgE
US6919366B2 (en) 1998-05-22 2005-07-19 Avanir Pharmaceuticals Benzimidazole derivatives as modulators of IgE
KR100336090B1 (en) 1998-06-27 2002-05-27 윤승원 Solid dispersed preparation of poorly water-soluble drug containing oil, fatty acid or mixture thereof
US6387885B1 (en) 1998-08-26 2002-05-14 Abbott Laboratories 3′,3′-N-bis-desmethyl-3′-N-cycloalkyl erythromycin derivatives as LHRH antagonists
US6248363B1 (en) 1999-11-23 2001-06-19 Lipocine, Inc. Solid carriers for improved delivery of active ingredients in pharmaceutical compositions
DE19913692A1 (en) 1999-03-25 2000-09-28 Basf Ag Mechanically stable pharmaceutical dosage forms containing liquid or semi-solid surface-active substances
ES2245955T3 (en) 1999-12-21 2006-02-01 Sugen, Inc. 7-AZA-INDOLIN-2-ONAS 4-SUBSTITUTES AND THEIR USE AS PROTEIN-KINASE INHIBITORS.
EP1268525B1 (en) 2000-04-05 2008-12-31 Schering Corporation Macrocyclic ns3-serine protease inhibitors of hepatitis c virus comprising n-cyclic p2 moieties
DE10026698A1 (en) 2000-05-30 2001-12-06 Basf Ag Self-emulsifying active ingredient formulation and use of this formulation
US7065453B1 (en) 2000-06-15 2006-06-20 Accelrys Software, Inc. Molecular docking technique for screening of combinatorial libraries
ES2281434T3 (en) 2000-08-14 2007-10-01 Ortho-Mcneil Pharmaceutical, Inc. REPLACED PIRAZOLS.
US6388093B1 (en) 2001-01-05 2002-05-14 Sun Chemical Corporation Syntheses for preparing 1,4-diketopyrrolo [3,4-c]pyrroles
PL208713B1 (en) 2001-06-11 2011-05-31 Virochem Pharma Inc Thiophene derivatives as antiviral agents for flavivirus infection
EP2335700A1 (en) 2001-07-25 2011-06-22 Boehringer Ingelheim (Canada) Ltd. Hepatitis C virus polymerase inhibitors with a heterobicylic structure
RU2286343C2 (en) 2001-08-10 2006-10-27 Орто-Макнейл Фармасьютикал, Инк. Substituted pyrazoles
US6828301B2 (en) 2002-02-07 2004-12-07 Boehringer Ingelheim International Gmbh Pharmaceutical compositions for hepatitis C viral protease inhibitors
TW200304820A (en) 2002-03-25 2003-10-16 Avanir Pharmaceuticals Use of benzimidazole analogs in the treatment of cell proliferation
JP3925265B2 (en) 2002-03-25 2007-06-06 コニカミノルタホールディングス株式会社 Organic electroluminescence element and display device using the same
BR0311784A (en) 2002-06-14 2005-03-08 Merck & Co Inc Compound, pharmaceutical composition, methods of treating or preventing cancer, modulating mitotic spindle formation and inhibiting mitotic kinesin and process for manufacturing a pharmaceutical composition
JP4570955B2 (en) 2002-07-09 2010-10-27 バーテクス ファーマスーティカルズ インコーポレイテッド Imidazoles with protein kinase inhibitory activity
AU2003261434A1 (en) 2002-08-12 2004-02-25 Bristol-Myers Squibb Company Iminothiazolidinones as inhibitors of hcv replication
GB0229518D0 (en) 2002-12-19 2003-01-22 Astrazeneca Ab Chemical compounds
TW200508224A (en) 2003-02-12 2005-03-01 Bristol Myers Squibb Co Cyclic derivatives as modulators of chemokine receptor activity
RS20050741A (en) 2003-04-02 2007-06-04 Boehringer Ingelheim International Gmbh., Pharmaceutical compositions for hepatitis c viral protease inhibitors
US8025899B2 (en) 2003-08-28 2011-09-27 Abbott Laboratories Solid pharmaceutical dosage form
MXPA06004723A (en) 2003-10-27 2006-07-05 Vertex Pharma Combinations for hcv treatment.
BRPI0401908A (en) 2004-06-04 2006-01-17 Univ Rio De Janeiro Serine protease inhibitor compounds, process for obtaining and using for treatment of flaviviroses
US20060089385A1 (en) 2004-06-08 2006-04-27 Yong Cui Pharmaceutical compositions
US7772271B2 (en) 2004-07-14 2010-08-10 Ptc Therapeutics, Inc. Methods for treating hepatitis C
CN101027303A (en) 2004-07-27 2007-08-29 吉里德科学公司 Imidazo 4,5-dpyrimidines, their uses and methods of preparation
RU2007109207A (en) 2004-08-13 2008-09-20 Прикис Фамэсьютикэлс, Инк. (US) COMPOUNDS-MODULATORS OF Sphingosine-1-Phosphate Receptor Activity (OPTIONS), PHARMACEUTICAL COMPOSITION CONTAINING THE INDICATED COMPOUNDS, AND METHOD FOR TREATMENT OF DISORDERS, ASSOCIATED SECTION-1-Associated
TW200633718A (en) 2004-12-16 2006-10-01 Applied Research Systems Treatment of hepatitis c in the asian population
KR20070107062A (en) 2005-02-01 2007-11-06 와이어쓰 AMINO-PYRIDINES AS INHIBITORS OF beta;-SECRETASE
CN101146769A (en) 2005-02-14 2008-03-19 惠氏公司 Azolylacylguanidines as beta-secretase inhibitors
GB2438802A (en) 2005-02-28 2007-12-05 Univ Rockefeller Structure of the hepatitis C virus NS5A protein
US8143288B2 (en) 2005-06-06 2012-03-27 Bristol-Myers Squibb Company Inhibitors of HCV replication
PE20070099A1 (en) 2005-06-30 2007-02-06 Janssen Pharmaceutica Nv N-HETEROARYLPIPERAZINYL UREAS AS MODULATORS OF FATTY ACID AMIDE HYDROLASE
WO2007011284A1 (en) 2005-07-15 2007-01-25 Astrazeneca Ab Therapeutic agents
KR20080080170A (en) 2005-12-12 2008-09-02 제네랩스 테크놀로지스, 인코포레이티드 N-(5-membered heteroaromatic ring)-amido anti-viral compounds
EP1976829A2 (en) 2005-12-12 2008-10-08 Genelabs Technologies, Inc. N-(6-membered aromatic ring)-amido anti-viral compounds
CA2633760A1 (en) 2005-12-21 2007-07-05 Abbott Laboratories Anti-viral compounds
WO2007076034A2 (en) 2005-12-21 2007-07-05 Abbott Laboratories Anti-viral compounds
WO2007081517A2 (en) 2005-12-21 2007-07-19 Abbott Laboratories Anti-viral compounds
WO2007073405A1 (en) 2005-12-21 2007-06-28 Decode Genetics Ehf N-linked aryl heteroaryl inhibitors of lta4h for treating inflammation
WO2007082554A1 (en) 2006-01-23 2007-07-26 Istituto Di Ricerche Di Biologia Molecolare P Angeletti Spa Modulators of hcv replication
TW200812611A (en) 2006-03-20 2008-03-16 Vertex Pharma Pharmaceutical compositions
US8163792B2 (en) 2006-05-16 2012-04-24 Pharmascience Inc. IAP BIR domain binding compounds
JP2007320925A (en) 2006-06-02 2007-12-13 Mitsubishi Chemicals Corp Triarylamine-based compound and electrophotographic photosensitizer using the same and image-forming device
JP5198439B2 (en) 2006-06-08 2013-05-15 イーライ リリー アンド カンパニー Novel MCH receptor antagonist
BRPI0713025A2 (en) 2006-06-16 2012-04-17 Syngenta Participations Ag ethenyl carboxamide-derived compounds useful as microbiocides, method of controlling or preventing plant infestation useful by phytopathogenic microorganisms and composition for controlling and preventing said microorganisms
EP1880715A1 (en) 2006-07-19 2008-01-23 Abbott GmbH & Co. KG Pharmaceutically acceptable solubilizing composition and pharmaceutical dosage form containing same
WO2008014238A2 (en) 2006-07-24 2008-01-31 Tetralogic Pharmaceuticals Corporation Dimeric iap inhibitors
WO2008014236A1 (en) 2006-07-24 2008-01-31 Tetralogic Pharmaceuticals Corporation Dimeric iap inhibitors
US8329159B2 (en) 2006-08-11 2012-12-11 Bristol-Myers Squibb Company Hepatitis C virus inhibitors
US7745636B2 (en) 2006-08-11 2010-06-29 Bristol-Myers Squibb Company Hepatitis C virus inhibitors
US20100158862A1 (en) 2006-08-11 2010-06-24 Bristol-Myers Squibb Company Hepatitis C Virus Inhibitors
US8303944B2 (en) 2006-08-11 2012-11-06 Bristol-Myers Squibb Company Hepatitis C virus inhibitors
US7759495B2 (en) 2006-08-11 2010-07-20 Bristol-Myers Squibb Company Hepatitis C virus inhibitors
US7659270B2 (en) 2006-08-11 2010-02-09 Bristol-Myers Squibb Company Hepatitis C virus inhibitors
WO2008064218A2 (en) 2006-11-21 2008-05-29 Smithkline Beecham Corporation Amido anti-viral compounds
TW200831084A (en) 2006-11-21 2008-08-01 Genelabs Tech Inc Anti-viral compounds
EP2094276A4 (en) 2006-12-20 2011-01-05 Abbott Lab Anti-viral compounds
WO2008074450A2 (en) 2006-12-20 2008-06-26 Nicox S.A. Non-peptidic renin inhibitors nitroderivatives
BRPI0810178A2 (en) 2007-04-12 2014-09-23 Joyant Pharmaceuticals Inc SMAC MIME DIMERS AND THERMALS USEFUL AS ANTICCAN AGENTS
US7741347B2 (en) 2007-05-17 2010-06-22 Bristol-Myers Squibb Company Hepatitis C virus inhibitors
WO2009003009A1 (en) 2007-06-26 2008-12-31 Enanta Pharmaceuticals, Inc. Substituted pyrrolidine as anti-infectives
CN101796040A (en) 2007-07-06 2010-08-04 吉里德科学公司 The modulators of pharmacokinetic properties of therapeutical agent
EP2185154A2 (en) 2007-08-03 2010-05-19 Schering Corporation Method of treating cxcr3 mediated diseases using heterocyclic substituted piperazines
US8629171B2 (en) 2007-08-08 2014-01-14 Bristol-Myers Squibb Company Crystalline form of methyl ((1S)-1-((25)-2-(5-(4'-(2-((25)-1((2S)-2-((methoxycarbonyl)amino)-3-methylbutanoyl)-2-pyrrolidinyl)-1H-imidazol-2-yl)-1-pyrrolidinyl)carbonyl)-2-methylpropyl)carbamate dihydrochloride salt
US7728027B2 (en) 2007-08-08 2010-06-01 Bristol-Myers Squibb Company Process for synthesizing compounds useful for treating hepatitis C
CL2008003092A1 (en) 2007-10-19 2009-11-27 Abbott Gmbh & Co Kg Solid dispersion product comprising an active agent derived from n-aryl urea, a matrix-forming agent selected from cyclodextrins, polymers, lipids and a non-ionic surfactant; process for preparing said dispersion product; and a pharmaceutical dosage form comprising it.
GB0801199D0 (en) 2008-01-23 2008-02-27 Acal Energy Ltd Fuel cells
EP2250160B1 (en) 2008-01-25 2015-11-11 Millennium Pharmaceuticals, Inc. Thiophenes and their use as phosphatidylinositol 3-kinase (pi3k) inhibitors
CA2715400A1 (en) 2008-02-12 2009-08-20 Bristol-Myers Squibb Company Heterocyclic derivatives as hepatitis c virus inhibitors
BRPI0822335A2 (en) 2008-02-12 2019-09-24 Bristol-Myers Squibb Company hepatitis c virus inhibitors
SI2242752T1 (en) 2008-02-13 2013-03-29 Bristol-Myers Squibb Company Imidazolyl biphenyl imidazoles as hepatitis c virus inhibitors
US8147818B2 (en) 2008-02-13 2012-04-03 Bristol-Myers Squibb Company Hepatitis C virus inhibitors
US7704992B2 (en) 2008-02-13 2010-04-27 Bristol-Myers Squibb Company Hepatitis C virus inhibitors
US20100021540A1 (en) 2008-02-28 2010-01-28 Abbott Laboratories Tablets and Preparation Thereof
BRPI0911260A2 (en) 2008-04-15 2015-09-29 Intermune Inc compound, pharmaceutical composition, method of inhibiting ns3 / ns4 protease activity in vitro, and uses of compounds
US20110117081A1 (en) 2008-05-05 2011-05-19 Aegera Therapeutics, Inc. Functionalized pyrrolidines and use thereof as iap inhibitors
WO2009143361A1 (en) 2008-05-22 2009-11-26 Smithkline Beecham Corporation Amido anti-viral compounds
EP2310402A1 (en) 2008-06-27 2011-04-20 Aegera Therapeutics Inc. Bridged secondary amines and use thereof as iap bir domain binding compounds
RS53420B (en) 2008-07-22 2014-12-31 Msd Italia S.R.L. Combinations of a macrocyclic quinoxaline compound which is an hcv ns3 protease inhibitor with other hcv agents
NZ590550A (en) 2008-08-02 2013-05-31 Genentech Inc Inhibitors of Apoptosis (IAP) for treating cancer
US20120009141A1 (en) 2008-08-07 2012-01-12 Pharmascience Inc. Functionalized pyrrolidines and use thereof as iap inhibitors
US7906655B2 (en) 2008-08-07 2011-03-15 Bristol-Myers Squibb Company Hepatitis C virus inhibitors
UY32099A (en) 2008-09-11 2010-04-30 Enanta Pharm Inc HEPATITIS C SERINA PROTEASAS MACROCYCLIC INHIBITORS
US8383094B2 (en) 2008-10-01 2013-02-26 Bristol-Myers Squibb Company Hepatitis C virus inhibitors
US8946289B2 (en) 2008-11-19 2015-02-03 Duke University Manassatin compounds and methods of making and using the same
JP2010126571A (en) 2008-11-26 2010-06-10 Toyo Ink Mfg Co Ltd Organic electroluminescent element material and organic electroluminescent element
WO2010062821A1 (en) 2008-11-28 2010-06-03 Glaxosmithkline Llc Anti-viral compounds, compositions, and methods of use
US8865756B2 (en) 2008-12-03 2014-10-21 Presidio Pharmaceuticals, Inc. Inhibitors of HCV NS5A
BRPI0922366B8 (en) 2008-12-03 2021-05-25 Presidio Pharmaceuticals Inc compound, pharmaceutical composition and use of a compound
JP2012513410A (en) 2008-12-23 2012-06-14 アボット・ラボラトリーズ Antiviral compounds
RU2505540C2 (en) 2008-12-23 2014-01-27 Эббви Инк. Antiviral compounds
WO2010091413A1 (en) 2009-02-09 2010-08-12 Enanta Pharmaceuticals, Inc. Linked dibenzimidazole derivatives
US8314135B2 (en) 2009-02-09 2012-11-20 Enanta Pharmaceuticals, Inc. Linked dibenzimidazole antivirals
US8188132B2 (en) 2009-02-17 2012-05-29 Enanta Pharmaceuticals, Inc. Linked dibenzimidazole derivatives
US8242156B2 (en) 2009-02-17 2012-08-14 Enanta Pharmaceuticals, Inc. Linked dibenzimidazole derivatives
US8394968B2 (en) 2009-02-17 2013-03-12 Bristol-Myers Squibb Company Hepatitis C virus inhibitors
TWI438200B (en) 2009-02-17 2014-05-21 必治妥美雅史谷比公司 Hepatitis c virus inhibitors
US8420686B2 (en) 2009-02-17 2013-04-16 Enanta Pharmaceuticals, Inc. Linked diimidazole antivirals
US8637561B2 (en) 2009-02-17 2014-01-28 Enanta Pharmaceuticals, Inc. Linked diimidazole derivatives
WO2010096462A1 (en) 2009-02-17 2010-08-26 Enanta Pharmaceuticals, Inc Linked diimidazole derivatives
EP2398474A4 (en) 2009-02-23 2012-12-05 Presidio Pharmaceuticals Inc Inhibitors of hcv ns5a
WO2010094977A1 (en) 2009-02-23 2010-08-26 Arrow Therapeutics Limited Novel biphenyl compounds useful for the treatment of hepatitis c
US8426458B2 (en) 2009-02-27 2013-04-23 Enanta Pharmaceuticals, Inc. Hepatitis C Virus inhibitors
US8101643B2 (en) 2009-02-27 2012-01-24 Enanta Pharmaceuticals, Inc. Benzimidazole derivatives
US8673954B2 (en) 2009-02-27 2014-03-18 Enanta Pharmaceuticals, Inc. Benzimidazole derivatives
KR101411889B1 (en) 2009-02-27 2014-06-27 이난타 파마슈티칼스, 인코포레이티드 Hepatitis c virus inhibitors
US8507522B2 (en) 2009-03-06 2013-08-13 Enanta Pharmaceuticals, Inc. Hepatitis C virus inhibitors
US9150554B2 (en) 2009-03-27 2015-10-06 Presidio Pharmaceuticals, Inc. Fused ring inhibitors of hepatitis C
UA108351C2 (en) 2009-03-27 2015-04-27 Hepatitis C Virus Replication Inhibitors
CA2756255A1 (en) 2009-03-27 2010-09-30 Presidio Pharmaceuticals, Inc. Substituted bicyclic hcv inhibitors
US8796466B2 (en) 2009-03-30 2014-08-05 Bristol-Myers Squibb Company Hepatitis C virus inhibitors
US20110237636A1 (en) 2009-03-30 2011-09-29 Bristol-Myers Squibb Company Hepatitis C Virus Inhibitors
TWI476190B (en) 2009-03-30 2015-03-11 必治妥美雅史谷比公司 Hepatitis c virus inhibitors
CN102388046A (en) 2009-04-08 2012-03-21 巴斯夫欧洲公司 Pyrrolopyrrole derivatives, their manufacture and use as semiconductors
TW201038559A (en) 2009-04-09 2010-11-01 Bristol Myers Squibb Co Hepatitis C virus inhibitors
US8143414B2 (en) 2009-04-13 2012-03-27 Bristol-Myers Squibb Company Hepatitis C virus inhibitors
MX2011010905A (en) 2009-04-15 2011-11-01 Abbott Lab Anti-viral compounds.
MX2011011136A (en) 2009-04-24 2011-11-18 Tibotec Pharm Ltd Diaryl ethers.
WO2010132538A1 (en) 2009-05-12 2010-11-18 Schering Corporation Fused tricyclic aryl compounds useful for the treatment of viral diseases
NZ619205A (en) 2009-05-13 2015-04-24 Gilead Pharmasset Llc Antiviral compounds
US8138215B2 (en) 2009-05-29 2012-03-20 Bristol-Myers Squibb Company Hepatitis C virus inhibitors
WO2010138791A1 (en) 2009-05-29 2010-12-02 Schering Corporation Antiviral compounds composed of three linked aryl moieties to treat diseases such as hepatitis c
US8211928B2 (en) 2009-05-29 2012-07-03 Bristol-Myers Squibb Company Hepatitis C virus inhibitors
CA2762885A1 (en) 2009-05-29 2010-12-02 Schering Corporation Antiviral compounds composed of three aligned aryl moieties to treat diseases such as hepatitis c
JP5530514B2 (en) 2009-06-11 2014-06-25 アッヴィ・バハマズ・リミテッド Antiviral compounds for treating HCV infection
US8716454B2 (en) 2009-06-11 2014-05-06 Abbvie Inc. Solid compositions
US8937150B2 (en) 2009-06-11 2015-01-20 Abbvie Inc. Anti-viral compounds
US8221737B2 (en) 2009-06-16 2012-07-17 Enanta Pharmaceuticals, Inc. Hepatitis C virus inhibitors
WO2010148006A1 (en) 2009-06-16 2010-12-23 Enanta Pharmaceuticals, Inc. Hepatitis c virus inhibitors
US8232246B2 (en) 2009-06-30 2012-07-31 Abbott Laboratories Anti-viral compounds
US8609648B2 (en) 2009-07-02 2013-12-17 Enanta Pharmaceuticals, Inc. Hepatitis C virus inhibitors
WO2011004276A1 (en) 2009-07-06 2011-01-13 Pfizer Limited Hepatitis c virus inhibitors
US8354419B2 (en) 2009-07-16 2013-01-15 Vertex Pharmaceuticals Incorporated Benzimidazole analogues for the treatment or prevention of flavivirus infections
RU2540897C2 (en) 2009-08-07 2015-02-10 Тиботек Фармасьютикалз Bis-benzimidazole derivatives as hepatitis c virus inhibitors
US20120172368A1 (en) 2009-09-03 2012-07-05 Koen Vandyck Bis-Benzimidazole Derivatives
US8344155B2 (en) 2009-09-04 2013-01-01 Glaxosmith Kline Llc Chemical compounds
US8822700B2 (en) 2009-09-11 2014-09-02 Enanta Pharmaceuticals, Inc. Hepatitis C virus inhibitors
US8927709B2 (en) 2009-09-11 2015-01-06 Enanta Pharmaceuticals, Inc. Hepatitis C virus inhibitors
US8703938B2 (en) 2009-09-11 2014-04-22 Enanta Pharmaceuticals, Inc. Hepatitis C virus inhibitors
US8815928B2 (en) 2009-09-11 2014-08-26 Enanta Pharmaceuticals, Inc. Hepatitis C virus inhibitors
EP2475256A4 (en) 2009-09-11 2013-06-05 Enanta Pharm Inc Hepatitis c virus inhibitors
EP2475254A4 (en) 2009-09-11 2013-05-22 Enanta Pharm Inc Hepatitis c virus inhibitors
US8759332B2 (en) 2009-09-11 2014-06-24 Enanta Pharmaceuticals, Inc. Hepatitis C virus inhibitors
US8415374B2 (en) 2009-10-12 2013-04-09 Bristol-Myers Squibb Company Combinations of hepatitis C virus inhibitors
WO2011050146A1 (en) 2009-10-23 2011-04-28 Glaxosmithkline Llc Chemical compounds
UA108211C2 (en) 2009-11-04 2015-04-10 Янссен Рід Айрленд Benzimidazole imidazole derivatives
US20110269956A1 (en) 2009-11-11 2011-11-03 Bristol-Myers Squibb Company Hepatitis C Virus Inhibitors
US20110274648A1 (en) 2009-11-11 2011-11-10 Bristol-Myers Squibb Company Hepatitis C Virus Inhibitors
US20110281910A1 (en) 2009-11-12 2011-11-17 Bristol-Myers Squibb Company Hepatitis C Virus Inhibitors
US20120276047A1 (en) 2009-11-25 2012-11-01 Rosenblum Stuart B Fused tricyclic compounds and derivatives thereof useful for the treatment of viral diseases
CN102482269B (en) 2009-12-04 2015-07-29 财团法人卫生研究院 Proline derivative
EP2512480A4 (en) 2009-12-14 2013-05-15 Enanta Pharm Inc Hepatitis c virus inhibitors
US8377980B2 (en) 2009-12-16 2013-02-19 Bristol-Myers Squibb Company Hepatitis C virus inhibitors
WO2011075607A1 (en) 2009-12-18 2011-06-23 Intermune, Inc. Novel inhibitors of hepatitis c virus replication
SG181797A1 (en) 2009-12-18 2012-07-30 Idenix Pharmaceuticals Inc 5,5-fused arylene or heteroarylene hepatitis c virus inhibitors
US20130156731A1 (en) 2009-12-22 2013-06-20 Kevin X. Chen Fused tricyclic compounds and methods of use thereof for the treatment of viral diseas
AU2010336355A1 (en) 2009-12-24 2012-07-05 Vertex Pharmaceuticals Incorporated Analogues for the treatment or prevention of flavivirus infections
US8362020B2 (en) 2009-12-30 2013-01-29 Bristol-Myers Squibb Company Hepatitis C virus inhibitors
WO2011091446A1 (en) 2010-01-22 2011-07-28 Glaxosmithkline Llc Chemical compounds
AU2011207492A1 (en) 2010-01-25 2012-08-16 Enanta Pharmaceuticals, Inc. Hepatitis C virus inhibitors
WO2011091532A1 (en) 2010-01-28 2011-08-04 Boehringer Ingelheim International Gmbh Hepatitis c inhibitor compounds
US8178531B2 (en) 2010-02-23 2012-05-15 Enanta Pharmaceuticals, Inc. Antiviral agents
CA2791630A1 (en) 2010-03-04 2011-09-09 Enanta Pharmaceuticals, Inc. Combination pharmaceutical agents as inhibitors of hcv replication
PE20130062A1 (en) 2010-03-09 2013-02-28 Merck Sharp & Dohme FUSED TRICYCLIC SILILO COMPOUNDS FOR THE TREATMENT OF VIRAL DISEASES
UA104517C2 (en) 2010-03-10 2014-02-10 Ебботт Леборетріз Solid composition useful for treating hepatitis c infection and method for its manufacturing
WO2011119870A1 (en) 2010-03-24 2011-09-29 Vertex Pharmaceuticals Incorporated Analogues for the treatment or prevention of flavivirus infections
WO2011119853A1 (en) 2010-03-24 2011-09-29 Vertex Pharmaceuticals Incorporated Analogues for the treatment or prevention of flavivirus infections
TW201141857A (en) 2010-03-24 2011-12-01 Vertex Pharma Analogues for the treatment or prevention of flavivirus infections
TW201139438A (en) 2010-03-24 2011-11-16 Vertex Pharma Analogues for the treatment or prevention of flavivirus infections
WO2011127350A1 (en) 2010-04-09 2011-10-13 Enanta Pharmaceuticals, Inc. Hepatitis c virus inhibitors
US20110312996A1 (en) 2010-05-17 2011-12-22 Intermune, Inc. Novel inhibitors of hepatitis c virus replication
EP2575475A4 (en) 2010-05-28 2013-11-27 Presidio Pharmaceuticals Inc Inhibitors of hcv ns5a
US8778938B2 (en) 2010-06-04 2014-07-15 Enanta Pharmaceuticals, Inc. Hepatitis C virus inhibitors
US20130310427A1 (en) 2010-06-09 2013-11-21 Presidio Pharmaceuticals, Inc. Inhibitors of hcv ns5a protein
WO2011156757A1 (en) 2010-06-10 2011-12-15 Gilead Sciences, Inc. Combination of anti-hcv compounds with ribavirin for the treatment of hcv
NZ605440A (en) 2010-06-10 2014-05-30 Abbvie Bahamas Ltd Solid compositions comprising an hcv inhibitor
EP2603080A4 (en) 2010-08-12 2014-01-22 Enanta Pharm Inc Hepatitis c virus inhibitors
US8822520B2 (en) 2010-09-22 2014-09-02 Presidio Pharmaceuticals, Inc. Substituted bicyclic HCV inhibitors
CN103249730A (en) 2010-09-24 2013-08-14 百时美施贵宝公司 Hepatitis c virus inhibitors
WO2012050848A1 (en) 2010-09-29 2012-04-19 Schering Corporation Fused tetracycle derivatives and methods of use thereof for the treatment of viral diseases
US20130280214A1 (en) 2010-09-29 2013-10-24 Merck Sharp & Dohme Corp. Polycyclic heterocycle derivatives and methods of use thereof for the treatment of viral diseases
CA2811662A1 (en) 2010-09-29 2012-04-05 Merck Sharp & Dohme Corp. Tetracyclic indole derivatives for treating hepatitis c virus infection
WO2012040923A1 (en) 2010-09-29 2012-04-05 Merck Sharp & Dohme Corp. Tetracyclic indole derivatives and methods of use thereof for the treatment of viral diseases
WO2012040924A1 (en) 2010-09-29 2012-04-05 Merck Sharp & Dohme Corp. Fused tetracyclic heterocycle compounds and methods of use thereof for treatment of viral diseases
EP2621932A4 (en) 2010-09-29 2014-03-26 Merck Sharp & Dohme Tetracyclic heterocycle compounds for treating hepatitis c viral infection
EP2635571B1 (en) 2010-11-04 2015-08-12 Theravance Biopharma R&D IP, LLC Novel inhibitors of hepatitis c virus
AU2011328980B2 (en) 2010-11-17 2015-07-30 Gilead Pharmasset Llc Antiviral compounds
RU2452735C1 (en) 2010-11-30 2012-06-10 Александр Васильевич Иващенко Substituted azoles, antiviral active component, pharmaceutical composition, synthesis and application method
WO2012087976A2 (en) 2010-12-21 2012-06-28 Intermune, Inc. Novel inhibitors of hepatitis c virus replication
TW201446286A (en) 2013-01-31 2014-12-16 Gilead Pharmasset Llc Solid dispersion formulation of an antiviral compound

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2010017432A1 (en) * 2008-08-07 2010-02-11 Schering Corporation Pharmaceutical formulations of an hcv protease inhibitor in a solid molecular dispersion
WO2010097229A2 (en) * 2009-02-27 2010-09-02 Ortho-Mcneil-Janssen Pharmaceuticals Inc Amorphous salt of a macrocyclic inhibitor of hcv
US20120172290A1 (en) * 2009-06-11 2012-07-05 Abbott Laboratories Anti-Viral Compounds

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
Liu, R. Water-Insoluble Drug Formulation, Second Edition; CRC Press, 2008, pages 499-503; 8 pages. *

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN113209087A (en) * 2020-02-05 2021-08-06 歌礼药业(浙江)有限公司 Pharmaceutical composition for inhibiting coronavirus and application thereof

Also Published As

Publication number Publication date
US9034832B2 (en) 2015-05-19

Similar Documents

Publication Publication Date Title
CA2857339C (en) Solid compositions comprising an hcv inhibitor
EP2579854B2 (en) Solid compositions
US8716454B2 (en) Solid compositions
US9034832B2 (en) Solid compositions
US9044480B1 (en) Compositions and methods for treating HCV
AU2014200725B2 (en) Solid compositions

Legal Events

Date Code Title Description
AS Assignment

Owner name: ABBVIE INC., ILLINOIS

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:GAO, YI;ZHANG, GEOFF;SIGNING DATES FROM 20141114 TO 20141117;REEL/FRAME:034802/0666

STCF Information on status: patent grant

Free format text: PATENTED CASE

MAFP Maintenance fee payment

Free format text: PAYMENT OF MAINTENANCE FEE, 4TH YEAR, LARGE ENTITY (ORIGINAL EVENT CODE: M1551); ENTITY STATUS OF PATENT OWNER: LARGE ENTITY

Year of fee payment: 4

MAFP Maintenance fee payment

Free format text: PAYMENT OF MAINTENANCE FEE, 8TH YEAR, LARGE ENTITY (ORIGINAL EVENT CODE: M1552); ENTITY STATUS OF PATENT OWNER: LARGE ENTITY

Year of fee payment: 8